From: [email protected] To: [email protected] Subject: Condition Petition for Joseph Jeffries, RPh Date: Wednesday, December 30, 2020 2:16:28 PM

This message was sent from the Condition page on medicalmarijuana.ohio.gov.

Box was check regarding file size being too large to upload. Action needed!

Name: Joseph Jeffries, RPh Address: 1865 Dresden Ave, East Liverpool, OH, 43920 Phone: (740) 391-2331 Email: [email protected]

Specific Disease or Condition: Panic Disorder with Agoraphobia

Information from experts who specialize in the disease or condition. The College of Family Physicians of Canada (CFPC) has created a guidance document for the use of cannabis in the treatment of anxiety. They cite the research of Drs Meldon Kahan, Anita Srivastava, Sheryl Spithoff, and Lisa Bromley. (see attached)

Relevant medical or scientific evidence pertaining to the disease or condition. Panic Disorder affects 6M adults in the US(2.7% of the population)and is characterized by sudden attacks of intense fear or anxiety, usually associated with ...symptoms such as heart palpitations, rapid breathing or SOB, blurred vision, dizziness, and racing thoughts. Often these symptoms are thought to be a heart attack by the individual, and many cases are diagnosed in hospital emergency rooms.” Cost est. at $33billion (see attached)

Consideration of whether conventional medical therapies are insufficient to treat or alleviate the disease or condition. Anxiety disorders are treated with psychotherapy, medications or both. Conventional medications used include benzodiazepines, antidepressants, and beta-blockers. BZDs have been implicated in a lg. % of opioid-related overdose deaths In long-term users cessation of BZDs can result in a withdrawal syndrome, manifesting in anxiety, seizures, and sleep disorders. BZD related deaths rose from 1,135 in 1999 to 11,537 in 2018 according to the National Institute on Drug Abuse (see attached)

Evidence supporting the use of medical marijuana to treat or alleviate the disease or condition, including journal articles, peer-reviewed studies, and other types of medical or scientific documentation. *Cannabinoid related agents in the treatment of anxiety disorders. Discusses the modulation of anxiety responses to herbal, synthetic and endogenous cannabinoids. *Naturalistic examination of the perceived effects on negative affects. Nearly 12,000 tracked sessions were analyzed to determine changes in depression, anxiety and stress. *Cannabidiol as a potential treatment for anxiety disorders. (see all attached) Letters of support provided by physicians with knowledge of the disease or condition. This may include a letter provided by the physician treating the petitioner, if applicable. Letter from Vincent Paolone, MD Diplomat American Board of Psychiatry and Neurology since 1997 and Fellow of the American Psychiatric Association since 2014 (see attached) THE COLLEGE OF LE COLLÈGE DES FAMILY PHYSICIANS MÉDECINS DE FAMILLE OF CANADA DU CANADA

Authorizing Dried Cannabis for Chronic Pain or Anxiety

preliminary guidance

September 2014 This guidance document was prepared on behalf of the College of Family Physicians of Canada (CFPC) by members of the Addiction Medicine and Chronic Pain Program Committees, in collaboration with members of the Child and Adolescent Health, Maternity and Newborn Care, Mental Health, Palliative Care, and Respiratory Medicine Program Committees, of CFPC’s Section of Family Physicians with Special Interests or Focused Practices.

Copyright © 2014 by College of Family Physicians of Canada

Suggested citation College of Family Physicians of Canada. Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance from the College of Family Physicians of Canada. Mississauga, ON: College of Family Physicians of Canada; 2014.

Disclaimer This document has been prepared by the CFPC to provide preliminary, rather than comprehensive, guidance, based on what is currently known about the use of cannabis for certain medical purposes. Dried cannabis is not an approved drug or medicine in Canada, and the provision of this information should not be interpreted as an endorsement of the use of this product, or of cannabis generally, by the CFPC.

The content within this document is provided for information and education purposes about a new and largely unstudied area of clinical practice. It is not intended to substitute for the advice of a physician. Patients should always consult their doctors for specific information on personal health matters, or other relevant professionals to ensure that their own circumstances are considered.

The CFPC accepts no responsibility or liability arising from any error or omission or from the use of any information contained herein.

Reproduction of the CFPC logo or hyperlinking to this document for commercial purposes is strictly prohibited.

Contact us The College of Family Physicians of Canada welcomes your feedback.

We are working to ensure that the recommendations in this guidance document continue to reflect the latest available evidence and to incorporate the practice expertise of CFPC members who use them.

If you have suggestions for additions or changes to this document, we would appreciate receiving them. All feedback received will be considered for inclusion in the revised guide, to be released Winter 2015.

Please forward your suggestions to [email protected].

The College of Family Physicians of Canada 2630 Skymark Avenue Mississauga ON L4W 5A4 Telephone: (905) 629-0900 Email: [email protected]

Find us on the Web: www.cfpc.ca

ii Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Contents

Introduction...... iv Table 3. Clinical features of cannabis use disorder in patients with chronic pain...... 11 Methods...... 1 Assessment, monitoring, and discontinuation...... 12 How to navigate this document...... 2 recommendation 8...... 12 A. Summary of Recommendations...... 3 recommendation 9...... 12 General principles...... 3 Strategies to prevent harm...... 13 Misuse prevention and intervention...... 4 recommendation 10...... 13 Assessment, monitoring, and discontinuation...... 4 recommendation 11...... 13 Strategies to prevent harm...... 4 Communication with patients and consultants...... 14 Communication with patients and consultants...... 4 recommendation 12...... 14 Dosing...... 5 Table 4. CAGE-AID Tool...... 14 B. Discussion and Supporting Evidence...... 5 Table 5. Advice for patients about safety General principles...... 5 and harm reduction...... 14 recommendation 1...... 5 Table 6. Messages to patients who disagree with recommendation 2...... 5 your decision to not authorize cannabis treatment...... 15

recommendation 3...... 6 recommendation 13...... 15

recommendation 4...... 6 Dosing...... 16

recommendation 5...... 7 recommendation 14...... 16

recommendation 6...... 8 Table 7. Strains containing 9% THC or less, by licensed producer*...... 18 Table 1. Provincial regulatory authorities’ policies on authorizing dried cannabis...... 9 recommendation 15...... 19

Table 2. Sample treatment agreement...... 10 Conclusions...... 19

Misuse prevention and intervention...... 11 Acknowledgements...... 20

recommendation 7...... 11 References...... 21

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance iii Introduction The Health Canada Marihuana for Medical Purposes Regulations (MMPR),1 which came into force on April 1, 2014, permit a physician to sign a medical document authorizing a patient’s access to, and the dispensing of, a specified quantity of the dried cannabis plant, which patients purchase through a licensed producer. The medical document has a format and function similar to a prescription. However, dried cannabis differs from prescribed products in that Health Canada has not reviewed data on its safety or effectiveness and has not approved it for therapeutic use.

This situation puts family physicians in a difficult position: we are asked to authorize our patients’ access to a product with little evidence to support its use, and in the absence of regulatory oversight and approval.

To address this predicament, this document offers family physicians preliminary guidance on the authorizing of dried cannabis for chronic pain or anxiety, pending the development of formal guidelines. Although the MMPR speak only of use for medical purposes without specifying any diagnoses, the writing group chose chronic pain and anxiety as the clinical areas to highlight because they may be the most common conditions for which a patient requests authorization for cannabis from a family physician.

Research shows that dried cannabis is a potent, psychoactive substance that can have significant acute and chronic cognitive effects. Its acute effects include perceptual distortions, cognitive impairment, euphoria, and anxiety.2 Chronic use of dried cannabis may be associated with persistent neuropsychological deficits, even after a period of abstinence.3,4 The patient may report initial benefit from smoking cannabis and, as with many mood-altering substances such as alcohol, opioids, benzodiazepines, and cocaine, experience temporary relief from pain or anxiety. However, these products have the potential to cause harm by impairing memory and cognition, worsening performance at work and school, and by interfering with social relationships. Before authorizing cannabis, family physicians need to consider if there is sufficient evidence that the anticipated therapeutic benefits for the patient’s particular health condition outweigh the potential harms. Similarly, continuation of the cannabis is warranted only if the authorizing physician is satisfied that there has been improvement in the patient’s pain level, function, and/or quality of life, and that there are no signs that the patient is at risk of misuse or harm.

iv Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Methods This document was written by members of the Addiction Medicine and Chronic Pain Program Committees of the Section of Family Physicians with Special Interests or Focused Practices (SIFP) of the College of Family Physicians of Canada (CFPC), in collaboration with other individuals and SIFP Program Committees: Child and Adolescent Health, Maternity and Newborn Care, Mental Health, Palliative Care, and Respiratory Medicine. CFPC Program Committees are made up of members with a special interest, and often enhanced expertise, in a specific clinical domain that is relevant to the practice of family medicine.

The writing team based the document on a literature search and review of evidence on specific topics related to cannabis effectiveness, safety, and adverse effects. The team acknowledges the research of Kahan and colleagues,5 which has been adapted in the preparation of this document. The material appears with the permission of the publisher, Canadian Family Physician.

Members of the participating program committees collaborated to prepare a succession of drafts, which then underwent review by an editorial team. A subgroup of the editorial team wrote the final document on behalf of the participants. The final document was taken to the entire group for its consensus before publication.

Recommendations were graded as Level I (based on well-conducted controlled trials or meta-analyses), Level II (well-conducted observational studies), or Level III (expert opinion; for the purposes of this document, consensus among the committee members drafting this document on behalf of the CFPC).

The context within which the participants worked to produce this document is extraordinary, as we have described above: authorization of a largely unstudied substance, particularly challenging medical practice areas (pain and addiction), intense interest from patients (often accompanied by less interest in evidence), an absence of regulation, and, above all, an urgency to provide basic parameters to guide family physicians in the safe treatment of their patients.

The individuals named in the Acknowledgements agreed to be listed as contributors on the basis that this document:

• Was urgently needed to address a knowledge gap in a controversial practice area without the usual supports and • Provides preliminary guidance while the CFPC engages in a rigorous process to provide more formal clinical practice guidelines and continuing professional development offerings

Terminology Medical marijuana: This term is in popular use but is imprecise, referring broadly to dried cannabis dispensed or otherwise obtained and used either for supervised medical purposes or for self-medication. In a scientific context we prefer to use the term “dried cannabis.”

Dried cannabis/cannabis: We use these terms interchangeably to refer to the substance under discussion in this paper: the product that a patient may purchase through a licensed producer, under the MMPR, if he or she has a medical document authorizing its dispensing.

Pharmaceutical cannabinoids: This term refers to the prescription drugs nabilone (Cesamet) and nabiximols (Sativex). Marinol (dronabinol) was previously available but has been removed from the Canadian market by the manufacturer.

Medical document: Health Canada uses this term to denote the prescription-like form that physicians complete and sign to authorize patients’ access to dried cannabis from licensed producers. Health Canada provides a sample medical document on its website.6

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 1 How to navigate this document This document is organized into two parts. The first, “A. Summary of Recommendations,” outlines the recommendations in brief, sketching in point form the new and still developing landscape within which family physicians find themselves regarding “medical marijuana”:

• The federal regulations that give the physician the responsibility for granting access to this unregulated substance • The as-yet limited evidence regarding cannabis’s effects and efficacy in clinical use • The degree to which evidence derived from studies of pharmaceutical cannabinoids can be applied to dried cannabis, and vice versa • The provincial medical regulatory authorities’ requirements of physicians regarding signing medical documents for cannabis • The issues and questions that arise in the sometimes challenging conversations between physicians and patients surrounding cannabis

The second part, “B. Discussion and Supporting Evidence,” provides a fuller discussion of these topics. It describes:

• What we know to date about the potential harms and benefits of cannabis use in various populations and for treating different conditions, with a focus on pain and anxiety • Regulations and suggested best practices to follow before authorizing and continuing a patient’s access to cannabis

It also provides practical resources to use in clinical practice, including:

• Messages for patients • Tools to use when screening patients for misuse or addiction risk • A sample treatment agreement • Information about the strains available from licensed producers • Calculations for dosing

In sections A and B, the recommendations are grouped under the headings:

• General principles (recommendations 1–6) • Misuse prevention and intervention (recommendation 7) • Assessment, monitoring, and discontinuation (recommendations 8 and 9) • Strategies to prevent harm (recommendations 10 and 11) • Communication with patients and consultants (recommendations 12 and 13) • Dosing (recommendations 14 and 15)

2 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance A. Summary of Recommendations To navigate to the discussion and evidence for an individual recommendation, click on the hyperlinked heading.

General principles

Recommendation 1 There is no research evidence to support the authorization of dried cannabis as a treatment for pain conditions commonly seen in primary care, such as fibromyalgia or low back pain (Level III). Authorizations for dried cannabis should only be considered for patients with neuropathic pain that has failed to respond to standard treatments (Level I).

Recommendation 2 If considering authorizing dried cannabis for treatment of neuropathic pain, the physician should first consider a) adequate trials of other pharmacologic and nonpharmacologic therapies and b) an adequate trial of pharmaceutical cannabinoids (Level I).

Recommendation 3 Dried cannabis is not an appropriate therapy for anxiety or insomnia (Level II).

Recommendation 4 Dried cannabis is not appropriate for patients who:

a) Are under the age of 25 (Level II) b) Have a personal history or strong family history of psychosis (Level II) c) Have a current or past cannabis use disorder (Level III) d) Have an active substance use disorder (Level III) e) Have cardiovascular disease (angina, peripheral vascular disease, cerebrovascular disease, arrhythmias) (Level III) f) Have respiratory disease (Level III) or g) Are pregnant, planning to become pregnant, or breastfeeding (Level II)

Recommendation 5 Dried cannabis should be authorized with caution in those patients who:

a) Have a concurrent active mood or anxiety disorder (Level II) b) Smoke tobacco (Level II) c) Have risk factors for cardiovascular disease (Level III) or d) Are heavy users of alcohol or taking high doses of opioids or benzodiazepines or other sedating medications prescribed or available over the counter (Level III)

Recommendation 6 Physicians should follow the regulations of their provincial medical regulators when authorizing dried cannabis (Level III).

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 3 Misuse prevention and intervention

Recommendation 7 Physicians should assess and monitor all patients on cannabis therapy for potential misuse or abuse (Level III). Assessment, monitoring, and discontinuation

Recommendation 8 Before signing a medical document authorizing dried cannabis for pain, the physician should do all of the following:

a) Conduct a pain assessment (Level II) b) Assess the patient for anxiety and mood disorders (Level II) c) Screen and assess the patient for substance use disorders (Level II)

Recommendation 9 The physician should regularly monitor the patient’s response to treatment with dried cannabis, considering the patient’s function and quality of life in addition to pain relief (Level III). The physician should discontinue authorization if the therapy is not clearly effective or is causing the patient harm (Level III). Strategies to prevent harm

Recommendation 10 Patients taking dried cannabis should be advised not to drive for at least:

a) Four hours after inhalation (Level II) b) Six hours after oral ingestion (Level II) c) eight hours after inhalation or oral ingestion if the patient experiences euphoria (Level II)

Recommendation 11 When authorizing dried cannabis therapy for a patient, the physician should advise the patient of harm reduction strategies (Level III). Communication with patients and consultants

Recommendation 12 The physician should manage disagreements with patients about decisions around authorization, dosing, or other issues with unambiguous, evidence-based statements (Level III).

Recommendation 13 The physician who is authorizing cannabis for a particular clinical indication must be primarily responsible for managing the care for that condition and following up with the patient regularly (Level III). Physicians seeking a second opinion on the potential clinical use of cannabis for their patient should only refer to facilities that meet standards for quality of care typically applied to specialized pain clinics (Level III). In both instances, it is essential that the authorizing physician, if not the patient’s most responsible health care provider, communicate regularly with the family physician providing ongoing comprehensive care for the patient (Level III).

4 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Dosing

Recommendation 14 Given the weak evidence for benefit and the known risks of using cannabis, the only sensible advice for physicians involved with authorizing dried cannabis is the maxim “Start low, and go slow” (Level III).

Recommendation 15 Although it is not required by the MMPR, physicians should specify the percentage of THC on the medical document for all authorizations for dried cannabis, just as they would specify dosing when prescribing any other analgesic (Level III).

B. Discussion and Supporting Evidence

General principles Recommendation 1 There is no research evidence to support the authorization of dried cannabis as a treatment for pain conditions commonly seen in primary care, such as fibromyalgia or low back pain (Level III). Authorizations for dried cannabis should only be considered for patients with neuropathic pain that has failed to respond to standard treatments (Level I).

To date, five controlled trials have examined dried cannabis in the treatment of chronic neuropathic pain.7-11 The trials were small, included patients who had previously smoked cannabis, and lasted from 1 to 15 days. Functional status, quality of life, and other important outcomes were not measured. No head-to-head comparisons of therapeutic benefits or adverse effects were made with other standard treatments for these conditions, or with pharmaceutical cannabinoid preparations.

The safety and effectiveness of dried cannabis has not been studied for pain conditions such as fibromyalgia and low back pain. No controlled studies have been conducted on dried cannabis for osteoarthritis, and the Canadian Rheumatology Association does not endorse the use of dried cannabis for either fibromyalgia or osteoarthritis.12 The oral pharmaceutical cannabinoid nabilone has some evidence of benefit for these conditions, although the evidence is weaker than for first-line treatments.13,14 Family physicians are advised to recommend other treatments with more evidence of safety and efficacy for these conditions.

Recommendation 2 If considering authorizing dried cannabis for treatment of neuropathic pain, the physician should first consider a) adequate trials of other pharmacologic and nonpharmacologic therapies and b) an adequate trial of pharmaceutical cannabinoids (Level I).

There are many pharmacologic and nonpharmacologic treatments documented as effective in the treatment of neuropathic pain, and these established therapies should be tried before moving on to trials of cannabinoids. Oral and buccal pharmaceutical cannabinoids have a larger body of evidence of efficacy than has dried cannabis in the treatment of neuropathic pain,15-20 although, apart from Sativex (indicated for neuropathic pain associated with multiple sclerosis or cancer), these drugs’ use for this treatment is off label. Evidence suggests that oral cannabinoids are also safer, with a lower risk of addiction and with milder cognitive effects.19,21-27

However, until further research is conducted, the same contraindications and precautions that apply to dried cannabis apply to pharmaceutical cannabinoids. Patients who request cannabis but refuse a trial of pharmaceutical cannabinoids may be using cannabis for euphoria rather than analgesia.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 5 Recommendation 3 Dried cannabis is not an appropriate therapy for anxiety or insomnia (Level II).

To our knowledge, there have been no controlled studies to date on the use of dried cannabis in the treatment of anxiety disorders. There is, however, a strong and consistent association between cannabis use and anxiety and mood disorders, although causality has not been established.28-38 Acute cannabis use can trigger anxiety and panic attacks,39 and studies on animals and human volunteers suggest that high doses of cannabis actually worsen anxiety.40 Cannabis use may worsen psychiatric impairment in patients with anxiety disorders.36,41,42

The tetrahydrocannabinol (THC) content of cannabis is associated with anxiety,41 though this relationship appears to be bidirectional.43,44 Physicians should consider the THC content of available cannabis and consider authorizing, if at all, only lower-strength strains for patients with anxiety.45 Regular users of cannabis might experience early symptoms of cannabis withdrawal, including an exacerbation of anxiety, when they abstain; withdrawal symptoms can ultimately be resolved through cannabis cessation.46

The evidence for using pharmaceutical cannabinoids in the treatment of anxiety and insomnia is stronger than the evidence for using dried cannabis. Small trials have demonstrated that oral nabilone improves sleep in patients with fibromyalgia or post-traumatic stress disorder.13,47 An oral extract of pure cannabidiol has been shown to relieve symptoms of social anxiety.48

Recommendation 4 Dried cannabis is not appropriate for patients who:

a) Are under the age of 25 (Level II) b) Have a personal history or strong family history of psychosis (Level II) c) Have a current or past cannabis use disorder (Level III) d) Have an active substance use disorder (Level III) e) Have cardiovascular disease (angina, peripheral vascular disease, cerebrovascular disease, arrhythmias) (Level III) f) Have respiratory disease (Level III) or g) Are pregnant, planning to become pregnant, or breastfeeding (Level II)

Patients under the age of 25 (Level II) Youth who smoke cannabis are at greater risk than older adults for cannabis-related psychosocial harms, including suicidal ideation, illicit drug use, cannabis use disorder, and long-term cognitive impairment.4,33,49-52

Patients with current, past, or strong family history of psychosis (Level II) Observational studies have demonstrated a consistent association between cannabis use in adolescence and persistent psychosis.53-60

Patients with current or past cannabis use disorder (Level III) Pain patients with cannabis use disorder should be counseled to discontinue cannabis and be referred for addiction treatment.

Patients with an active substance use disorder (Level III) Dried cannabis should not be authorized for any patient with a current problematic use of alcohol, opioids, or other drugs.

6 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Patients with cardiovascular disease (Level III) Cannabis use causes acute physiological effects such as hypertension, tachycardia, catecholamine release, and vascular constriction.61-64 There have been reports of young people suffering cardiovascular events shortly after smoking cannabis.65-67

Patients with respiratory disease (Level III) Heavy cannabis smoking may be an independent risk factor for impaired respiratory function and chronic obstructive pulmonary disease.68,69

Use of smoked cannabinoids has been found to increase the risk of airflow obstruction and hyperinflation but has been less associated with macroscopic emphysema.70 The cannabis use was associated with increased risk of lung cancer71 and head and neck cancer.72 The respiratory symptoms associated with dried cannabis use include wheezing apart from colds, exercise-induced shortness of breath, nocturnal wakening with chest tightness, and early morning sputum production.73

The depth of inhalation and the length of time the breath is held are usually greater when smoking marijuana than when smoking cigarettes. This means exposure to the chemicals in the smoke is greater for cannabis than for tobacco cigarettes, even though the frequency of smoking may be less. Cannabis smokers, for example, end up with five times more carbon monoxide in their bloodstream than do tobacco smokers.71

Patients who are pregnant, planning to become pregnant, or breastfeeding (Level II) Preliminary evidence links cannabis use during pregnancy to neurodevelopmental abnormalities in infants.74 Cannabis enters the breast milk and is contraindicated in women who are breastfeeding.

Recommendation 5 Dried cannabis should be authorized with caution in those patients who:

a) Have a concurrent active mood or anxiety disorder (Level II) b) Smoke tobacco (Level II) c) Have risk factors for cardiovascular disease (Level III) or d) Are heavy users of alcohol or taking high doses of opioids or benzodiazepines or other sedating medications prescribed or available over the counter (Level III)

Patients with current mood and anxiety disorders (Level II) Caution should be used when authorizing cannabis for patients with current mood or anxiety disorders, for the reasons outlined in Recommendation 3. If patients with co-existing anxiety and neuropathic pain are authorized for cannabis treatment, i) the dose should be kept low to avoid triggering anxiety, ii) the provider should consider indicating low THC-content or cannabidiol-only (CBD-only) strains on the medical document, and iii) cannabis should be discontinued if the patient’s anxiety or mood worsens.

Tobacco smokers (Level II) Even after controlling for tobacco smoking, cannabis smoking has been associated with lung cancer75 and chronic bronchitis. Patients who smoke tobacco should be strongly advised to use cannabis via vaporization rather than by smoking it.

Patients with risk factors for cardiovascular disease (Level II) Physicians are advised to use considerable caution when authorizing dried cannabis for use by patients with risk factors for cardiovascular disease (see Recommendation 4e). The dose should be kept low, and the patient should be encouraged to take it through vaporization or the oral route rather than by smoking it.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 7 Patients who are heavy users of alcohol, or taking high doses of opioids or benzodiazepines (Level III) Cannabis use can worsen the cognitive impairment caused by opioids, benzodiazepines, other sedatives, and alcohol.76 Patients taking dried cannabis should be advised to use alcohol in moderation, and physicians should consider tapering patients on high doses of opioids or benzodiazepines.77,78

Recommendation 6 Physicians should follow the regulations of their provincial medical regulators when authorizing dried cannabis (Level III).

Many of the provincial/territorial regulatory bodies have released policies on the authorization of cannabis.79 These regulators advise physicians to conduct a thorough assessment and to try conventional alternatives before providing a medical document for cannabis. Additional requirements, which vary considerably from province to province, are summarized below and in Table 1. Physicians should review the complete policy of their provincial regulator before signing a medical document for cannabis.

Conflict of interest Physicians must not have a financial interest in a company that produces medical marijuana, and they should follow their provincial regulatory authority’s Code of Ethics regarding potential conflicts of interest. Under the usual circumstances described in the MMPR, the licensed producer couriers the dried cannabis to the patient. Under extraordinary circumstances (if, for example, the patient does not have a postal address) the physician may receive and store dried cannabis. Consultation with provincial regulatory authorities about all such arrangements is advised.

Authorizations Several provinces require physicians to:

• State the patient’s medical condition on the medical document • Register with the regulator as a cannabis authorizer • Send the regulator a copy of the medical document, and/or keep the medical documents on a separate record for possible inspection

Some provinces specify that only the physician who manages the patient’s condition may write a medical document authorizing cannabis, so that the therapy occurs in the most potentially beneficial context of continuing and comprehensive care. An ongoing doctor-patient relationship is similarly important when visits are conducted using telemedicine—where patient and physician must communicate via an interface rather than face to face. For this reason, authorization of dried cannabis by physicians not usually involved in the patient’s care and using telemedicine is problematic; the authorizing physician is compelled to monitor response to treatment, emergence of adverse effects, and signs and symptoms of addiction without being physically present with the patient. This raises clear questions about whether care quality to these standards is possible in the context of a relationship that is carried out via telemedicine.

Documentation and consent Several regulators recommend that the patient sign a written treatment agreement (see Table 2), that the physician document that other treatments have been tried, and that the patient is aware of the risks of dried cannabis. They also recommend that the patient be reassessed at least every three months.

Assessment and monitoring for cannabis misuse Several provincial regulators advise physicians to use a standardized tool to assess the patient’s risk of addiction, and to have a procedure or protocol for identifying cannabis misuse.

8 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Table 1. Provincial regulatory authorities’ policies on authorizing dried cannabis Medical Regulatory Authorities Requirements Applying to Physicians (Provincial Colleges or Councils) Conflict of interest BC AB SK MB QC NB NL Must not apply to become a licensed producer ● ● Must not store, provide, or dispense marijuana ● ● ● ● Must not have any financial or management interest in a ● ● licensed distributer or producer Must not have any personal gain from providing a ● non-medical service Authorizations BC AB SK MB QC NB NL State patient’s medical condition on medical document ● ● ● Register with regulator as a dried cannabis authorizer ● Provide a copy of the medical document to the regulator ● ● Send original medical document to licensed producer, give ● copy to patient, and enter another copy in chart Review available prescription databases to determine ● ● patient’s medication usage Keep all medical documents on a separate record for ● ● ● inspection by the College May only sign a medical document authorizing cannabis for a patient if he or she is the primary manager of the ● ● ● patient’s medical condition May not authorize cannabis through telemedicine ● Keep a register of cannabis patients so they can be invited ● to participate in the research database projects Consent and documentation BC AB SK MB QC NB NL Inform the patient that cannabis can only be authorized as ● part of the research database project Ask the patient to read the patient information document ● Have patient sign a written treatment agreement ● ● Have the patient sign a written consent form ● ● Document that the patient was informed of the risks and ● ● ● ● ● benefits, and that other treatments were tried Assess the patient at least every three months ● ● Assessment and monitoring BC AB SK MB QC NB NL Complete the assessment and follow-up form available on ● the regulator’s website Cannabis misuse BC AB SK MB QC NB NL Assess the patient’s risk of addiction using standardized tool ● ● ● Have a process or protocol for identifying misuse ● ● ● Source: Canadian Consortium for the Investigation of Cannabinoids, www.ccic.net/index.php?id=248,703,0,0,1,0. Accessed 2014 Jun 17.

Please visit CFPC’s website for information on the latest statements and requirements from the provincial regulatory authorities: http://www.cfpc.ca/medical_marijuana/

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 9 Table 2. Sample treatment agreement Because we take our responsibilities to authorize and supervise the medical use of marijuana (dried cannabis) very seriously, we ask you to read, understand, and sign this form.

1. i request Dr ______, MD, to sign a medical document for me under the Health Canada MMPR legislation, so that I may legally use marijuana to treat my medical condition. 2. i agree to receive a medical document for marijuana only from one physician, Dr ______, MD. 3. i agree to consume no more marijuana than the doses authorized for me by Dr ______, MD. I will not request a refill before the agreed-upon refill date. 4. i agree to not distribute my marijuana to any other person, for personal use or for sale. I am aware that redistribution of any marijuana for sale is an illegal activity. 5. i am aware that using marijuana is associated with psychosis in persons who are still undergoing neurodevelopment (brain growth). Therefore, I will ensure that no person under the age of 25 years has access to my marijuana. 6. i agree to the safe storage of my marijuana. 7. i am aware that taking marijuana with other substances, especially sedating substances, may cause harm and possibly even death. I will not use illegal drugs (eg, cocaine, heroin) or controlled substances (eg, narcotics, stimulants, anxiety pills) that were not prescribed for me. 8. i will not use controlled substances that were prescribed by another doctor unless Dr ______, MD, is aware of this. 9. i agree to testing (eg, urine drug screening) when and as requested by my physician. 10. i agree to have an office visit and medical assessment at least every _____ (months or weeks). 11. i understand that Health Canada has provided access to marijuana by signed medical document from a physician for the treatment of certain medical conditions, but despite this, Health Canada has not approved marijuana as a registered medication in Canada. 12. i understand that my physician may not be knowledgeable about all of the risks associated with the use of a non-Health Canada- approved substance like marijuana. 13. i agree to communicate to my physician, Dr ______, MD, any experiences of altered mental status or possible medical side effects of the use of marijuana. 14. i accept full responsibility for any and all risks associated with the use of marijuana, including theft, altered mental status, and side effects of the product. 15. i am aware that marijuana use is not advisable during pregnancy and breastfeeding. I agree to inform my physician, Dr ______, MD, if I am pregnant. 16. i am aware that smoking any substance can cause harm and medical complications to my breathing and respiratory status. I will avoid smoking marijuana. I will avoid mixing marijuana with tobacco. I agree to use my marijuana only by vaporizer or as an edible product. 17. i am aware that my physician may discontinue authorizing marijuana for my condition if he or she assesses that the medical or mental health risk or side effects are too high. 18. i agree to see specialists or therapists about my condition at my physician’s request. 19. i agree to avoid driving a vehicle or operating heavy machinery for at least 4 hours after the use of marijuana, and for longer if I feel any persistent negative effects on my ability to drive. 20. As per the Health Canada MMPR legislation, I agree to purchase my marijuana only from a licensed producer. I am aware that possession of marijuana from other sources is illegal. 21. i am aware that any possible criminal activity related to my marijuana use may be investigated by legal authorities and criminal charges may be laid. During the course of an investigation, legal authorities have the right to access my medical information with a warrant. 22. Following the terms of this contract is one of the conditions I must meet to access marijuana for treatment. I understand that if I violate any of this agreement’s terms, my physician may stop authorizing my use of cannabis. 23. Dr ______, MD, has the right to discuss my health care issues with other health care professionals or family members if it is felt, on balance, that my safety outweighs my right to confidentiality.

Patient’s printed name Patient’s signature

Date Practitioner’s signature

10 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Misuse prevention and intervention Recommendation 7 Physicians should assess and monitor all patients on cannabis therapy for potential misuse or abuse (Level III).

All patients using dried cannabis regularly should be monitored carefully and assessed routinely for cannabis use disorder. Clinical features of cannabis use disorder are listed in Table 3. Patients with suspected cannabis use disorder should be advised that they will likely experience improved mood and better function if they stop or reduce their use. Patients who are unable to stop or reduce should be referred for formal addiction treatment. Cannabis should not be authorized for patients with current problematic use of cannabis, alcohol, or other drugs (see Recommendation 5d).

Before authorizing cannabis use for the patient, the physician should take a careful history of current and past substance use, including cannabis, alcohol, tobacco, prescription opioids, and benzodiazepines, and illicit drugs such as heroin and cocaine. Several medical regulatory authorities recommend using a standardized tool to assess the risk of addiction. The CAGE-AID instrument80 is one such simple tool (Table 4). A urine drug screen may also be included in the initial assessment.

If the patient does not use substances problematically and begins cannabis treatment, the physician should ask the patient at each office visit about cognitive and mood-altering effects, as well as compliance with the dosing recommendations and use of any other substances. Periodic urine drug screens are advised.

The authorization for cannabis should be discontinued if the patient:

• Runs out early or uses cannabis from other sources • Begins to use alcohol, opioids, or other drugs problematically • Begins to show signs of a cannabis use disorder

Table 3. Clinical features of cannabis use disorder in patients with chronic pain

• Insists on a medical document for dried cannabis rather than trying other treatments known to be effective for his or her condition • Uses cannabis daily or almost daily, spending considerable non-productive time on this activity • Has poor school, work, and social functioning • Is currently addicted to or misusing other substances (other than tobacco) • Has risk factors for cannabis use disorder: is young, has current mood or anxiety disorder or a history of addiction or misuse • Reports having difficulty stopping or reducing use • Reports cannabis withdrawal symptoms after a day or more of abstinence: intense anxiety, fatigue • Has friends or family members concerned about his or her cannabis use

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 11 Assessment, monitoring, and discontinuation Recommendation 8 Before signing a medical document authorizing dried cannabis for pain, the physician should do all of the following:

a) Conduct a pain assessment (Level II) b) Assess the patient for anxiety and mood disorders (Level II) c) Screen and assess the patient for substance use disorders (Level II)

The physician should ask the patient to rate the pain on a 10-point scale, and to describe the effect of the pain on daily activities, including sleep. The physician should also assess the patient’s mood. The physician should take a careful history of current and past substance use, including cannabis, alcohol, tobacco, prescription opioids and benzodiazepines, and illicit drugs such as heroin and cocaine. Several of the provincial medical regulators (the provincial licensing colleges) recommend a standardized tool to assess the risk of addiction; the CAGE-AID is one simple, validated tool81 available to physicians (Table 4). A urine drug screen is also advised, and the patient should be asked to read and sign a treatment agreement (Table 2).

Recommendation 9 The physician should regularly monitor the patient’s response to treatment with dried cannabis, considering the patient’s function and quality of life in addition to pain relief (Level III). The physician should discontinue authorization if the therapy is not clearly effective or is causing the patient harm (Level III).

At follow-up office visits, the physician should reassess the effects of cannabis on the patient’s pain ratings and function.

Many psychoactive drugs with abuse liability will temporarily blunt the patient’s perception of pain without improving function. All centrally acting analgesics can also cause sedation, euphoria, or cognitive impairment. To authorize or continue to authorize dried cannabis for the purpose of analgesia, physicians should be as certain as they would need to be in prescribing any other analgesic that its potential benefits are greater than its potential risks.

Dried cannabis therapy should be reassessed and possibly stopped in the following circumstances:

• The patient experiences insufficient analgesia and/or no improvement in function (note that some pain patients continue to complain of severe pain even as their function improves) • The treatment is not improving sleep, mood, function, and/or quality of life • The patient experiences side effects such as memory impairment, sedation, fatigue, and worsening functioning • The patient shows clinical features of cannabis use disorder (Table 3), such as running out early or using cannabis from other sources

12 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Strategies to prevent harm Recommendation 10 Patients taking dried cannabis should be advised not to drive for at least:

a) Four hours after inhalation (Level II) b) Six hours after oral ingestion (Level II) c) Eight hours after inhalation or oral ingestion if the patient experiences euphoria (Level II)

Cannabis use prior to driving is an independent risk factor for motor vehicle accidents.82-86 Patients should be advised not to drive for a minimum of four hours after inhalation or a minimum of six hours after oral ingestion87; they should abstain from driving for a full eight hours if they experience euphoria.88

However, note that “Health Canada states that the ability to drive or perform activities requiring alertness may be impaired for up to 24 hours following a single consumption.”12

Recommendation 11 When authorizing dried cannabis therapy for a patient, the physician should advise the patient of harm reduction strategies (Level III).

Some patients may consider dried cannabis to be “natural” and therefore safer than pharmaceutical products. They may be unaware that it is as important to follow dosing recommendations with dried cannabis as with any other course of treatment, and that different modes of delivery are safer or more precise than others.

For example, vaporization appears to be safer than smoking (combustion) as the vapour contains fewer toxic elements.89 Vaporization of herbal cannabis has also been evaluated in clinical trials.90 One such vaporizer is approved as a medical device in Canada (the Volcano Medic).91 However, long-term safety effects of unregulated cannabis vaporization techniques (such as e-cigarettes) are unknown at this time.

It is important to ensure that patients understand that potential side effects of cannabis, such as sedation or cognitive impairment, can impact their safety. As noted in Recommendation 10, Health Canada has stated that driving, operating heavy equipment, or other activities involving alertness and coordination may be unsafe for up to 24 hours following a single consumption, depending on the dosage, delivery route, and patient’s age and other health factors. It is important to discuss with patients that their reactions to the substance and to different formulations are individual, and that it is important to go slowly with the treatment until a stable, effective dose is reached.

We advise physicians to share patient education materials, such as the strategies in Table 5, with the patients they authorize for dried cannabis treatment.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 13 Communication with patients and consultants Recommendation 12 The physician should manage disagreements with patients about decisions around authorization, dosing, or other issues with unambiguous, evidence-based statements (Level III).

The main messages for the patient who requests cannabis are that a) cannabis is not an approved medicine and b) the medical literature to date reports little evidence of benefit and considerable risk of harm with its use (see Table 6).

Table 4. CAGE-AID Tool

Yes No ❏ ❏ Have you ever felt you ought to Cut down on your drinking or drug use? ❏ ❏ Have people Annoyed you by criticizing your drinking or drug use? ❏ ❏ Have you ever felt bad or Guilty about your drinking or drug use? ❏ ❏ Have you ever had a drink or used drugs first thing in the morning (Eye-opener) to steady your nerves, get rid of a hangover, or get the day started?

Scoring: One “positive” response indicates the need for further assessment. A urine drug screen (UDS) is also suggested. Source: Brown RL et al. Wis Med J 1995;94:135-140

Table 5. Advice for patients about safety and harm reduction

• Use the lowest dose necessary.

• Do not “breath hold” or take more cannabis than the dose your doctor has specified.

• We recommend you ingest (that is, eat) your cannabis or take it using a vaporizer instead of smoking it, to reduce your risk of exposure to toxins that result from burning the cannabis in a cigarette. This is important to help protect you from heart or lung disease.

• Do not use dried cannabis with alcohol or other sedating drugs.

• If you are smoking cannabis, do not mix tobacco into the cigarette.

• Do not give or sell your cannabis to others—it is both dangerous and illegal.

• Store your dried cannabis in a locked container, out of reach of children and hidden from visitors and from adolescents at home.

• Avoid smoking cannabis in your house, to limit exposure of family members to second-hand smoke.

• Do not drive for at least four hours after any use by any route, and for at least six hours after oral ingestion. Do not drive for at least eight hours after using cannabis if you experience euphoria when you use it.

• Do not use cannabis of any kind if you are pregnant or plan to become pregnant, or if you are breastfeeding.

14 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Table 6. Messages to patients who disagree with your decision to not authorize cannabis treatment

• Dried cannabis is not a good treatment for you, even if you experience less anxiety or pain right after use. Overall, it may be harming you. It can cause sedation and fatigue, depression, anxiety, or memory impairment. It can also interfere with your work, school, or social relationships.

• Dried cannabis has some serious risks and there is little evidence of benefit.

• Neither Health Canada nor any national medical organization has endorsed dried cannabis as a medicine. As a doctor I am bound to comply with the standards of my profession.

• We will work together to come up with an individualized treatment plan for you. Safe and effective treatments are available for your condition.

• If the patient is at high risk for cannabis-related harms, eg, is young or has a concurrent anxiety or substance use disorder: As your doctor I cannot prescribe any treatment that may harm you.

• If the patient refuses a trial of oral cannabinoids prior to any consideration of dried cannabis, explore the possibility that the patient is using dried cannabis for its effects on mood: If these drugs are not helping with pain relief or function, is it possible that you are getting a high from cannabis that makes it seem like it is helping pain for a while? If that’s so, the trouble is that the high can also impair your thinking and perception, which can create bigger problems for you.

• If the patient remains dissatisfied: I can’t authorize the use of an untested therapy when we have other, carefully studied and effective treatments that are safer and subject to strict quality control. I won’t authorize dried cannabis for you. I can refer you to a doctor who is a pain specialist, who can advise you on the risks and benefits of dried cannabis for your condition.

• If you suspect a cannabis use disorder: In my opinion, your use of cannabis could be causing you harm. We need to talk about ways to reduce or stop your cannabis use.

• If the patient says that your refusal forces him or her to purchase cannabis illegally: I advise you not to buy cannabis or any other drug from the street. In my opinion, using street cannabis is not benefiting your health, and it could be causing you harm.

Recommendation 13 The physician who is authorizing cannabis for a particular clinical indication must be primarily responsible for managing the care for that condition and following up with the patient regularly (Level III). Physicians seeking a second opinion on the potential clinical use of cannabis for their patient should only refer to facilities that meet standards for quality of care typically applied to specialized pain clinics (Level III). In both instances, it is essential that the authorizing physician, if not the patient’s most responsible health care provider, communicate regularly with the family physician providing ongoing comprehensive care for the patient (Level III).

Fragmentation of patient care is never advisable. Several regulatory authorities (see Recommendation 6) have advised that authorization of cannabis and care for a clinical condition that includes cannabis therapy should be managed by the most responsible health care provider for that patient.

Before referring a patient, the physician should first ensure that the clinic:

a) Has expertise in the patient’s medical or psychiatric condition b) routinely conducts a careful patient assessment prior to recommending any therapeutic intervention

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 15 c) Provides an explicit statement on the clinic’s policies on the indications, contraindications, and dosing for dried cannabis d) Does not have any financial conflicts of interest, such as charging patients fees, or financial involve- ment with licensed cannabis producers

The referring physician should send the consultant all clinically relevant information on the patient’s substance use, mental health, and pain history.

Dosing Recommendation 14 Given the weak evidence for benefit and the known risks of using cannabis, the only sensible advice for physicians involved with authorizing dried cannabis is the maxim “Start low, and go slow” (Level III).

The optimal dose should improve pain relief and function, while causing minimal euphoria or cognitive impairment. Gradual dose titration is needed to establish the dose’s effectiveness and safety. This is of critical importance because, as Health Canada has stated, even low doses of low-THC cannabis can cause cognitive impairment lasting as long as 24 hours in some individuals.2,12

What follows is a synthesis of what we know from the few controlled trials on dried cannabis available, and the medical literature on pharmaceutical cannabinoids. In the absence of rigorous evidence, we cannot overstress the importance of exhausting other possible therapies before embarking on a trial of cannabis therapy, as well as the necessity to “start low and go slow,” while continually monitoring the patient’s response to the treatment.

Suggested dosing: start low Determining a safe and effective dose for herbal cannabis is much more difficult than for pharmaceutical products, because individuals vary in their mode of administration (eg, inhaled versus oral), so that there can be a wide variation in the dose delivered. Wide interpatient dose variability is also noted for pharmaceutical cannabinoids.92

Subjects in one trial experienced relief of pain with one inhalation of 9.4% THC cannabis smoked three times per day. The single inhalation produced a serum level of 45 µg/L,11 a level slightly lower than the level associated with euphoria (50–100 µg/L).

Patients initiating cannabis therapy in inhaled form (smoked or vaporized) should start with very small amounts of herbal cannabis. Patients often measure their “dose” in terms of puffs; a single inhalation therefore represents a meaningful and intuitive “dose” form. Since the products available to the patient vary in the amount of cannabinoid they contain (cannabis strains have different cannabinoid profiles), by starting with a single inhalation of a new strain, the patient may slowly explore his or her response to the product. Starting with strains with lower THC levels is wise, because the lower percentage minimizes potential unwanted cognitive effects; higher doses of THC do not necessarily lead to better pain control.

Since medical documents need to specify 30-day quantities, and authorization takes effect on the date of signing, patients may order several grams over a one-month period; they may choose to purchase only a few grams of a given strain for two weeks, then to ask for a different strain. As long as they do not exceed the allowable 30-day limit, and are able to work with the licensed producer, patients may explore different THC and CBD profiles. The licensed producer may call the authorizing physician to confirm details of the authorization. We suggest requesting notification from the licensed producer whenever changes are made to what the physician has authorized (see Recommendation 15).

16 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance There are many reports of patients having to use larger quantities of herbal cannabis in the form of juicing (ie, maceration in a blender with liquids) or to prepare oral products; we simply do not have enough information to support these claims.

The following calculations are offered as preliminary pharmacokinetic considerations, based on several assumptions as outlined.

The amount of active cannabinoids delivered to the patient using herbal cannabis will depend on several factors, including the cannabinoid content of the source material and the mode of administration, as well as genetic and metabolic patient factors. Clearly the first two factors may be amenable to adjustment; the THC and CBD level of the herbal material is standardized by the licensed producers under the MMPR and physicians should suggest that patients begin with lower THC levels. The MMPR currently only allow for patients to receive dried herbal cannabis, and not any form of extract or oral edible product, so patients must also choose the mode of administration. Here the physician faces difficult choices; the inhaled route may be by vaporization, about which we have limited information, or by smoking, which is clearly not ideal but remains the most common means of cannabis self-administration.

It is useful to consider some broad considerations of these cannabis inhalation techniques to guide these discussions and decisions:

• Based on WHO estimates, an average “joint” contains 500 mg (0.5 g) of herbal cannabis. A typical tobacco cigarette, by comparison, weighs 1.0 g. • Studies of smoked cannabis for neuropathic pain conditions suggest effective doses ranging from one single inhalation from 25 mg of herbal cannabis containing 9.4% THC three times daily using a pipe,11 to 9 inhalations from a 900 mg “joint” of herbal cannabis containing 7% THC.7,8 This translates into current evidence for a daily inhaled dose of 100–700 mg of up to 9% THC content dried cannabis. • It is worth noting that in all studies the incidence of adverse events increases with increasing THC levels.

In the only study to date of vaporized cannabis for neuropathic pain, the amount of herbal material placed in the vaporizer was 800 mg and subjects took between 8 and 12 inhalations from the vaporizer balloon over a two- hour period.90 Once again, analgesic effects were noted at low THC levels and side effects increased with the THC level of administered cannabis.

Most studies of smoked or vaporized cannabis use a standardized inhalation procedure: inhale slowly over 5 seconds, hold breath for 10 seconds, then gently exhale.

Until further dose and delivery system information becomes available, these data may be crudely fashioned to provide patients with the following guidance and information:

1. They are advised to consider using vaporized cannabis over smoked cannabis. 2. They should use inhaled cannabis in a well-ventilated, private, and calm environment. 3. The authorization for dried cannabis will be for the lowest effective level of THC available. 4. They should start any new cannabis product with a slow single inhalation, and then wait four hours so that they can fully appreciate the effects. 5. They should allow for several single inhalation trials of a product to observe and then discuss their responses with their physicians, before either increasing the number of inhalations or changing their order with the producer. 6. As with all psychoactive drugs, they must be informed of and alert to cannabis’s potential mood-alter- ing, euphoric, or sedative effects, which can occur and present risk even at very low doses. 7. They should keep notes on effects and experiences throughout the therapy to facilitate discussion with their authorizing physician and other health professionals.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 17 Increasing dosage: go slow Although the MMPR allows physicians to authorize as much as 5.0 g of dried cannabis per patient per day, we expect that analgesic benefit will occur for most patients at considerably lower doses. We expect that the upper level to the safe use of dried cannabis will be on the order of 3.0 g per day, and that even this level of use should be considered only in very circumscribed conditions:

• This dosing level would apply to experienced users of dried cannabis only, never to cannabis-naïve patients • It must only be arrived at through a careful process of assessing the patient’s response as dosage is slowly increased, weighing analgesic benefit, improvement in function, and presence or absence of adverse effects

Furthermore, physicians considering authorizing dried cannabis at doses higher than the current evidence supports (an inhaled dose of 100–700 mg of no more than 9% THC cannabis daily) are strongly advised to:

• Discuss the decision to increase the dosage, either approaching or exceeding a 3.0 g/day level, with a trusted and experienced colleague • Document in the patient’s record the reasons that support the increased dosage

Table 7 lists the licensed producers in Canada, and the names of the strains they sell that contain 9% THC or less.93

Table 7. Strains containing 9% THC or less, by licensed producer* Company Variety and Percentage THC Bedrocan Bediol: 6.5% Bedrolite: 0.5% Canna Farms No strains 9% or less Cannimed 9.9: 9% Cannimed Cannimed 1.13: 0.7% Delta 9 biotech Does not list % THC on website In the Zone Does not list % THC on website Mettrum Purple #2: 7.9% Green #1, 5: 5.5% Green #2: 5.5% MedReleaf Corp Avidekel: 1.1% Organigram Not listed Peace Naturals Harvest Moon: 9% Nina: 8% Thunderbird Not listed Tilray No strains 9% or less Tweed Argyle: 5% Whistler No strains 9% or less

*Information compiled May 2014. Source: Health Canada, Authorised licensed producers under the MMPR, 2014

18 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance Recommendation 15 Although it is not required by the MMPR, physicians should specify the percentage of THC on all medical documents authorizing dried cannabis, just as they would specify dosing when prescribing any other analgesic (Level III).

The THC concentrations used in the five controlled trials on neuropathic pain (see Recommendation 1) ranged from 1% to 9%. Physicians should be aware that some commercial strains have THC concentrations as high as 15%–30%; these concentrations may increase the risk of cognitive impairment.

Therefore, the physician should note on the medical document to “Supply dried cannabis containing 9% THC or less. Send information on % THC composition directly to physician’s office. Notify physician of any change in THC concentration of product given to patient.”

The MMPR authorization document also requires indication of a daily quantity of cannabis. As indicated above (Recommendation 14), at present, the medical literature supports a daily dose of 100–700 mg.

Conclusions As stated earlier, the CFPC developed this guidance document in response to a clearly expressed need from members for assistance in navigating an extraordinary practice situation. They have been caught between their desire and obligation to provide evidence-informed care for their patients and a law that appears, to patients at least, to compel them to deal with dried cannabis as if it were a medicine.

For that reason, this document has been prepared with a sense of urgency. We are confident in the practice expertise and judgment of those members who participated in its creation, but recognize that the clinical conditions it deals with and the lack of solid evidence for almost any assertion in this area make giving clear- cut advice difficult. We have tried, nonetheless, to provide guidance that is as definitive as possible because we recognize that family physicians will not be able to avoid making decisions when their patients approach them about this topic.

We will, as an organization, continue to support efforts by Health Canada and other bodies to generate additional research evidence on the place of dried cannabis in the treatment of chronic pain, anxiety, and the variety of other conditions for which its use has been suggested. We encourage CFPC members to contact us, to add their input and share their experiences as we move forward safely and compassionately in this new and challenging area of therapeutics.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 19 Acknowledgements The team acknowledges the research of Drs Meldon Kahan, Anita Srivastava, Sheryl Spithoff, and Lisa Bromley5 used in the preparation of this guidance document. Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance from the College of Family Physicians of Canada is the result of many individuals’ collaborative efforts. All contributors listed below have reviewed and approved the final manuscript.

CFPC SIFP Program Committees The following program committees of the Section of Family Physicians with Special Interests or Focused Practices endorse the final manuscript.

Addiction Medicine Maternity and Newborn Care Palliative Care Child and Adolescent Health mental Health Respiratory Medicine Chronic Pain Core writing group The core writing group members contributed substantially to the document’s conception and design and/or research and analysis, drafted and reviewed the document for important intellectual content, and approved the final version to be published.

CFPC members Sharon Cirone, MD, CCFP(EM), FCFP; Chair, Addiction Medicine Program Committee Ruth E. Dubin, MD, PhD, FCFP, DAPPM, DCAPM; Chair, Chronic Pain Program Committee Meldon Kahan, MD, CCFP, FCFP; Member, Addiction Medicine Program Committee Mark A. Ware, MBBS, MRCP(UK), MSc

CFPC staff Jamie Meuser, MD, CCFP, FCFP; Executive Director, Professional Development and Practice Support Lynn Schellenberg, CPE; Writer/Editor Additional contributors The following contributors provided input on successive drafts and reviewed and approved the final manuscript.

CFPC members Alan Kaplan, MD, CCFP(EM), FCFP; Chair, Respiratory Medicine Program Committee Ellen Anderson, MD, MHSc; Chair, Mental Health Program Committee Lisa Graves, MD, CCFP, FCFP; Chair, Maternity and Newborn Care Program Committee Roxanne MacKnight, MD, CCFP, FCFP; Member, Child and Adolescent Health Program Committee Lori Montgomery, MD, CCFP; Member, Chronic Pain Program Committee Patricia Mousmanis, MD, CCFP, FCFP; Chair, Child and Adolescent Health Program Committee

CFPC staff Victor Ng, MSc, MD, CCFP(EM); Consulting Physician, Programs and Practice Support Roy Wyman, MD, CCFP, FCFP; Consulting Physician, Programs and Practice Support Financial disclosures Dr Ware has received research funding from Cannimed for clinical trials of vapourized cannabis for chronic pain disorders through the McGill University Health Centre Research Institute. Other competing interests None declared.

20 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance References

1. Canada. Marihuana for Medical Purposes Regulations, SOR/2013-119. Ottawa: Minister of Justice; 2014. Available from: http://laws-lois.justice.gc.ca/eng/regulations/SOR-2013-119/index.html. Accessed 2014 May 26. 2. Controlled Substances and Tobacco Directorate, Health Canada. Information for health care professionals: Cannabis (marihuana, marijuana) and the cannabinoids. Ottawa: Health Canada; 2013. Available from: www.hc-sc.gc.ca/dhp-mps/marihuana/med/infoprof-eng.php. Accessed 2014 May 13. 3. Gonzalez R. Acute and non-acute effects of cannabis on brain functioning and neuropsychological performance. Neuropsychol Rev 2007;17(3):347-361. 4. Meier MH, Caspi A, Ambler A, Harrington H, Houts R, Keefe RS, et al. Persistent cannabis users show neuropsychological decline from childhood to midlife. Proc Natl Acad Sci U S A 2012;109(40):E2657-2664. Doi: 10.1073/pnas.1206820109. Epub 2012 Aug 27. 5. Kahan M, Srivastava A, Spithoff S, Bromley L. Prescribing smoked cannabis for chronic noncancer pain. Preliminary recommendations. Can Fam Physician. In press. 6. Health Canada. Drugs and Health Products. Sample medical document for the Marihuana for Medical Purposes Regulations. Available from: www.hc-sc.gc.ca/dhp-mps/alt_formats/pdf/marihuana/info/med-eng.pdf. Accessed 2014 Jul 12. 7. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007;68(7):515-521. 8. Wilsey B, Marcotte T, Tsodikov A, Millman J, Bentley H, Gouaux B, et al. A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain 2008;9(6):506-521. Epub 2008 Apr 10. 9. Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009;34(3):672-680. Epub 2008 Aug 6. 10. Corey-Bloom J, Wolfson T, Gamst A, Jin S, Marcotte TD, Bentley H, et al. Smoked cannabis for spasticity in multiple sclerosis: a randomized, placebo-controlled trial. CMAJ 2012;184(10):1143-1150. 11. Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, et al. Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. CMAJ 2010;182(14):e694-e701. Epub 2010 Aug 30. 12. Fitzcharles MA, Clauw DJ, Ste-Marie PA, Shir Y. The dilemma of medical marijuana use by rheumatology patients. Arthritis Care Res (Hoboken) 2014;66(6):797-801. Doi: 10.1002/acr.22267. 13. Ware MA, Fitzcharles MA, Joseph L, Shir Y. The effects of nabilone on sleep in fibromyalgia: results of a randomized controlled trial. Anesth Analg 2010;110(2):604-610. Epub 2009 Dec 10. 14. Skrabek RQ, Galimova L, Ethans K, Perry D. Nabilone for the treatment of pain in fibromyalgia. J Pain 2008;9(2):164-173. 15. Barnes MP. Sativex: clinical efficacy and tolerability in the treatment of symptoms of multiple sclerosis and neuropathic pain. Expert Opin Pharmacother 2006;7(5):607-615. 16. Bestard JA, Toth CC. An open-label comparison of nabilone and gabapentin as adjuvant therapy or monotherapy in the management of neuropathic pain in patients with peripheral neuropathy. Pain Pract 2011;11(4):353-368. 17. Frank B, Serpell MG, Hughes J, Matthews JN, Kapur D. Comparison of analgesic effects and patient tolerability of nabilone and dihydrocodeine for chronic neuropathic pain: randomised, crossover, double blind study. BMJ 2008;336(7637):199-201. Epub 2008 Jan 8. 18. Toth C, Mawani S, Brady S, Chan C, Liu C, Mehina E, et al. An enriched-enrolment, randomized withdrawal, flexible-dose, double-blind, placebo-controlled, parallel assignment efficacy study of nabilone as adjuvant in the treatment of diabetic peripheral neuropathic pain. Pain 2012;153(10):2073-2082. 19. Aragona M, Onesti E, Tomassini V, Conte A, Gupta S, Gilio F, et al. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple sclerosis: a double-blind, placebo controlled, crossover study. Clin Neuropharmacol 2009;32(1):41-47. 20. Namaka M, Leong C, Grossberndt A, Klowak M, Turcotte D, Esfahani F, et al. A treatment algorithm for neuropathic pain: an update. Consult Pharm 2009;24(12):885-902. 21. Cooper ZD, Comer SD, Haney M. Comparison of the analgesic effects of dronabinol and smoked marijuana in daily marijuana smokers. Neuropsychopharmacology 2013;38(10):1984-1992. Doi: 10.1038/npp.2013.97. Epub 2013 Apr 22. 22. Mendelson JH, Mello NK. Reinforcing properties of oral delta 9-tetrahydrocannabinol, smoked marijuana, and nabilone: influence of previous marijuana use. Psychopharmacology (Berl) 1984;83(4):351-356. 23. Issa MA, Narang S, Jamison RN, Michna E, Edwards RR, Penetar DM, et al. The subjective psychoactive effects of oral dronabinol studied in a randomized, controlled crossover clinical trial for pain. Clin J Pain 2014;30(6):472-478. 24. Wesnes KA, Annas P, Edgar CJ, Deeprose C, Karlsten R, Philipp A, et al. Nabilone produces marked impairments to cognitive function and changes in subjective state in healthy volunteers. J Psychopharmacol 2010;24(11):1659-1669. Epub 2009 Jun 12.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 21 25. Kurzthaler I, Bodner T, Kemmler G, Entner T, Wissel J, Berger T, et al. The effect of nabilone on neuropsychological functions related to driving ability: an extended case series. Hum Psychopharmacol 2005;20(4):291-293. 26. Karschner EL, Darwin WD, McMahon RP, Liu F, Wright S, Goodwin RS, et al. Subjective and physiological effects after controlled Sativex and oral THC administration. Clin Pharmacol Ther 2011;89(3):400-407. Epub 2011 Feb 2. 27. Ware MA, St Arnaud-Trempe E. The abuse potential of the synthetic cannabinoid nabilone. Addiction 2010;105(3):494-503. 28. Saban A, Flisher AJ, Grimsrud A, Morojele N, London L, Williams DR, et al. The association between substance use and common mental disorders in young adults: results from the South African Stress and Health (SASH) Survey. Pan Afr Med J 2014;17(Suppl 1):11. 29. Crippa JA, Zuardi AW, Martín-Santos R, Bhattacharyya S, Atakan Z, McGuire P, et al. Cannabis and anxiety: a critical review of the evidence. Hum Psychopharmacol 2009;24(7):515-523. 30. Cheung JT, Mann RE, Ialomiteanu A, Stoduto G, Chan V, Ala-Leppilampi K, et al. Anxiety and mood disorders and cannabis use. Am J Drug Alcohol Abuse 2010;36(2):118-122. 31. Horwood LJ, Fergusson DM, Coffey C, Patton GC, Tait R, Smart D, et al. Cannabis and depression: an integrative data analysis of four Australasian cohorts. Drug Alcohol Depend 2012;126(3):369-378. 32. Kokkevi A, Richardson C, Olszewski D, Matias J, Monshouwer K, Bjarnason T. Multiple substance use and self-reported suicide attempts by adolescents in 16 European countries. Eur Child Adolesc Psychiatry 2012;21(8):443-450. 33. Fergusson DM, Horwood LJ, Swain-Campbell N. Cannabis use and psychosocial adjustment in adolescence and young adulthood. Addiction 2002;97(9):1123-1135. 34. Fergusson DM, Boden JM, Horwood LJ. Cannabis use and other illicit drug use: testing the cannabis gateway hypothesis. Addiction 2006;101(4):556-569. 35. Degenhardt L, Coffey C, Romaniuk H, Swift W, Carlin JB, Hall WD, et al. The persistence of the association between adolescent cannabis use and common mental disorders into young adulthood. Addiction 2013;108(1):124-133. 36. Buckner JD, Heimberg RG, Schneier FR, Liu SM, Wang S, Blanco C. The relationship between cannabis use disorders and social anxiety disorder in the National Epidemiological Study of Alcohol and Related Conditions (NESARC). Drug Alcohol Depend 2012;124(1-2):128-134. 37. Cougle JR, Bonn-Miller MO, Vujanovic AA, Zvolensky MJ, Hawkins KA. Posttraumatic stress disorder and cannabis use in a nationally representative sample. Psychol Addict Behav 2011;25(3):554-558. 38. Wong SS, Zhou B, Goebert D, Hishinuma ES. The risk of adolescent suicide across patterns of drug use: a nationally representative study of high school students in the United States from 1999 to 2009. Soc Psychiatry Psychiatr Epidemiol 2013;48(10):1611-1620. Epub 2013 Jun 7. 39. Dannon PN, Lowengrub K, Amiaz R, Grunhaus L, Kotler M. Comorbid cannabis use and panic disorder: short term and long term follow- up study. Hum Psychopharmacol 2004;19(2):97-101. 40. Moreira FA, Wotjak CT. Cannabinoids and anxiety. Curr Top Behav Neurosci 2010;2:429-450. 41. Lev-Ran S, Le Foll B, McKenzie K, Rehm J. Cannabis use and mental health-related quality of life among individuals with anxiety disorders. J Anxiety Disord 2012;26(8):799-810. 42. Arendt M, Rosenberg R, Fjordback L, Brandholdt J, Foldager L, Sher L, et al. Testing the self-medication hypothesis of depression and aggression in cannabis-dependent subjects. Psychol Med 2007;37(7):935-945. 43. Tambaro S, Bortolato M. Cannabinoid-related agents in the treatment of anxiety disorders: current knowledge and future perspectives. Recent Pat CNS Drug Discov 2012;7(1):25-40. 44. Ruehle S, Rey AA, Remmers F, Lutz B. The endocannabinoid system in anxiety, fear memory and habituation. J Psychopharmacol 2012;26(1):23-39. 45. Sarris J, McIntyre E, Camfield DA. Plant-based medicines for anxiety disorders, part 2: a review of clinical studies with supporting preclinical evidence [correction published in CNS Drugs 2013 Aug;27(8):675]. CNS Drugs 2013;27(4):301-319. 46. Budney AJ, Moore BA, Vaudrey RG, Hughes JR. The time course and significance of cannabis withdrawal. J Abnorm Psychol 2003;112(3):393-402. 47. Fraser GA. The use of a synthetic cannabinoid in the management of treatment-resistant nightmares in posttraumatic stress disorder (PTSD). CNS Neurosci Ther 2009;15(1):84-88. 48. Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology 2011;36(6):1219-1226. 49. Nocon A, Wittchen HU, Pfister H, Zimmermann P, Lieb R. Dependence symptoms in young cannabis users? A prospective epidemiological study. J Psychiatr Res 2006;40(5):394-403. Epub 2005 Sept 15. 50. Lynskey MT, Vink JM, Boomsma DI. Early onset cannabis use and progression to other drug use in a sample of Dutch twins. Behav Genet 2006;36(2):195-200.

22 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 51. Lisdahl KM, Price JS. Increased marijuana use and gender predict poorer cognitive functioning in adolescents and emerging adults. J Int Neuropsychol Soc 2012;18(4):678-688. Epub 2012 May 22. 52. Jager G, Ramsey NF. Long-term consequences of adolescent cannabis exposure on the development of cognition, brain structure and function: an overview of animal and human research. Curr Drug Abuse Rev 2008;1(2):114-123. 53. Hall W, Degenhardt L. Cannabis use and the risk of developing a psychotic disorder. World Psychiatry 2008;7(2):68-71. 54. Arseneault L, Cannon M, Witton J, Murray RM. Causal association between cannabis and psychosis: examination of the evidence. Br J Psychiatry 2004;184:110-117. 55. Moore TH, Zammit S, Lingford-Hughes A, Barnes TR, Jones PB, Burke M, et al. Cannabis use and risk of psychotic or affective mental health outcomes: a systematic review. Lancet 2007;370(9584):319-328. 56. Dragt S, Nieman DH, Schultze-Lutter F, van der Meer F, Becker H, de Haan L, et al. Cannabis use and age at onset of symptoms in subjects at clinical high risk for psychosis. Acta Psychiatr Scand 2012;125(1):45-53. Epub 2011 Aug 29. 57. Manrique-Garcia E, Zammit S, Dalman C, Hemmingsson T, Andreasson S, Allebeck P. Cannabis, schizophrenia and other non-affective psychoses: 35 years of follow-up of a population-based cohort. Psychol Med 2012;42(6):1321-1328. Epub 2011 Oct 17. 58. Le Bec PY, Fatséas M, Denis C, Lavie E, Auriacombe M. [Cannabis and psychosis: search of a causal link through a critical and systematic review.]. Review. French.]. Encephale 2009;35(4):377-385. Epub 2008 Jul 9. 59. Kuepper R, van Os J, Lieb R, Wittchen HU, Höfler M, Henquet C. Continued cannabis use and risk of incidence and persistence of psychotic symptoms: 10 year follow-up cohort study. BMJ 2011;342:d738. Doi: 10.1136/bmj.d738. 60. Davis GP, Compton MT, Wang S, Levin FR, Blanco C. Association between cannabis use, psychosis, and schizotypal personality disorder: findings from the National Epidemiologic Survey on Alcohol and Related Conditions. Schizophr Res 2013;151(1-3):197-202. Epub 2013 Nov 5. 61. Thomas G, Kloner RA, Rezkalla S. Adverse cardiovascular, cerebrovascular, and peripheral vascular effects of marijuana inhalation: what cardiologists need to know. Am J Cardiol 2014;113(1):187-190. 62. Sheikh HU, Mathew PG. Reversible cerebral vasoconstriction syndrome: updates and new perspectives. Curr Pain Headache Rep 2014;18(5):414. 63. Wolff V, Lauer V, Rouyer O, Sellal F, Meyer N, Raul JS, et al. Cannabis use, ischemic stroke, and multifocal intracranial vasoconstriction: a prospective study in 48 consecutive young patients. Stroke 2011;42(6):1778-1780. Epub 2011 Apr 21. 64. GW Pharma Ltd. Sativex product monograph. Last updated 2012 March 30. Available from: www.bayer.ca/files/SATIVEX-PM-ENG-30MAR2012-149598.pdf?#. Accessed 2014 Jul 12. 65. Hartung B, Kauferstein S, Ritz-Timme S, Daldrup T. Sudden unexpected death under acute influence of cannabis. Forensic Sci Int 2014;237:e11-e13. 66. Rodríguez-Castro CE, Alkhateeb H, Elfar A, Saifuddin F, Abbas A, Siddiqui T. Recurrent myopericarditis as a complication of Marijuana use. Am J Case Rep 2014;15:60-62. 67. Deharo P, Massoure PL, Fourcade L. Exercise-induced acute coronary syndrome in a 24-year-old man with massive cannabis consumption. Acta Cardiol 2013;68(4):425-428. 68. Reid PT, Macleod J, Robertson JR. Cannabis and the lung. J R Coll Physicians Edinb 2010;40(4):328-333. 69. Hall W, Degenhardt L. Adverse health effects of non-medical cannabis use. Lancet 2009;374(9698):1383-1391. 70. Aldington S, Williams M, Nowitz M, Weatherall M, Pritchard A, McNaughton A, Robinson G, Beasley R. Effects of cannabis on pulmonary structure, function and symptoms. Thorax 2007;62:1058-1063. 71. Aldington S, Harwood M, Cox B, Weatherall M, Beckert L, Hansell A, et al. Cannabis and Respiratory Disease Research Group. Cannabis use and risk of lung cancer: a case-control study. Eur Respir J 2008;31:280-286. 72. Aldington S, Harwood M, Cox B, Weatherall M, Beckert L, Hansell A, et al. Cannabis and Respiratory Disease Research Group. Cannabis use and cancer of the head and neck: case-control study. Otolaryngol Head Neck Surg 2008;138(3):374-380. 73. Taylor DR, Poulton R, Moffitt T, Ramankutty P, Sears M. The respiratory effects of cannabis dependence in young adults. Addiction 2000;95(11):1669–1677. 74. Wong S, Ordean A, Kahan M; Maternal Fetal Medicine Committee; Family Physicians Advisory Committee; Society of Obstetricians and Gynaecologists of Canada. Substance use in pregnancy. J Obstet Gynaecol Can 2011;33(4):367-384. 75. Callaghan RC, Allebeck P, Sidorchuk A. Marijuana use and risk of lung cancer: a 40-year cohort study. Cancer Causes Control 2013;24(10):1811-1820. Doi: 10.1007/s10552-013-0259-0. Epub 2013 Jul 12. 76. Sewell RA, Poling J, Sofuoglu M. The effect of cannabis compared with alcohol on driving. Am J Addict 2009;18(3):185-193. 77. Canadian Guideline for Safe and Effective Use of Opioids for Chronic Non-Cancer Pain. Canada: National Opioid Use Guideline Group (NOUGG); 2010. Available from: http://nationalpaincentre.mcmaster.ca/opioid/. Accessed 2014 May 13.

Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance 23 78. Reisfield GM. Medical cannabis and chronic opioid therapy. J Pain Palliat Care Pharmacother 2010;24(4):356-361. 79. Canadian Consortium for the Investigation of Cannabinoids. Provincial statements. www.ccic.net/index.php?id=248,703,0,0,1,0. Accessed 2014 Jun 17. 80. Brown RL, Rounds LA. Conjoint screening questionnaires for alcohol and other drug abuse: criterion validity in a primary care practice. Wis Med J 1995;94:135-140. 81. Couwenbergh C, Van Der Gaag RJ, Koeter M, De Ruiter C, Van den Brink W. Screening for substance abuse among adolescents: validity of the CAGE-AID in youth mental health care. Subst Use Misuse 2009;44(6):823-834. 82. Mann RE, Adlaf E, Zhao J, Stoduto G, Ialomiteanu A, Smart RG, et al. Cannabis use and self-reported collisions in a representative sample of adult drivers. J Safety Res 2007;38(6):669-674. Epub 2007 Nov 13. 83. Drummer OH, Gerostamoulos J, Batziris H, Chu M, Caplehorn J, Robertson MD, et al. The involvement of drugs in drivers of motor vehicles killed in Australian road traffic crashes. Accid Anal Prev 2004;36(2):239–248. 84. Laumon B, Gadegbeku B, Martin JL, Biecheler MB, SAM Group. Cannabis intoxication and fatal road crashes in France: population based case-control study [correction published in BMJ 2006;332(7553):1298]. BMJ 2005;331(7529):1371. Epub 2005 Dec 1. 85. Ramaekers JG, Berghaus G, van Laar M, Drummer OH. Dose related risk of motor vehicle crashes after cannabis use. Drug Alcohol Depend 2004;73(2):109-119. 86. Hartman RL, Huestis MA. Cannabis effects on driving skills. Clin Chem 2013;59(3):478-492. 87. Fischer B, Jeffries V, Hall W, Room R, Goldner E, Rehm J. Lower Risk Cannabis Use Guidelines for Canada (LRCUG): a narrative review of evidence and recommendations. Can J Public Health 2011;102(5):324-327. 88. Neavyn MJ, Blohm E, Babu KM, Bird SB. Medical marijuana and driving: a review. [Epub ahead of print.] J Med Toxicol 2014. 89. Abrams DI, Vizoso HP, Shade SB, Jay C, Kelly ME, Benowitz NL. Vaporization as a smokeless cannabis delivery system: a pilot study. Clin Pharmacol Ther 2007;82(5):572-578. 90. Wilsey B, Marcotte T, Deutsch R, Gouaux B, Sakai S, Donaghe H. Low-dose vaporized cannabis significantly improves neuropathic pain. J Pain 2013;14(2):136-148. 91. Canadian Consortium for the Investigation of Cannabinoids. Vaporization: approved medical devices. The Volcano Medic Vaporizer. www.ccic.net/index.php?id=132,744,0,0,1,0. Accessed 2014 Jul 31. 92. Serpell MG, Notcutt W, Collin C. Sativex long-term use: an open-label trial in patients with spasticity due to multiple sclerosis. J Neurol 2013;260(1):285-295. 93. Health Canada. Drugs and Health Products. List of authorised licensed producers under the Marihuana for Medical Purposes Regulations. Date modified 2014 Apr 22. Available from: www.hc-sc.gc.ca/dhp-mps/marihuana/info/list-eng.php. Accessed 2014 Jul 17.

24 Authorizing Dried Cannabis for Chronic Pain or Anxiety: Preliminary Guidance University of Tennessee Health Science Center UTHSC Digital Commons

Theses and Dissertations (ETD) College of Graduate Health Sciences

12-2013 Cost of Illness Study of Anxiety Disorders for the Ambulatory Adult Population of the United States Elaheh Shirneshan University of Tennessee Health Science Center

Follow this and additional works at: https://dc.uthsc.edu/dissertations Part of the Health and Medical Administration Commons

Recommended Citation Shirneshan, Elaheh , "Cost of Illness Study of Anxiety Disorders for the Ambulatory Adult Population of the United States" (2013). Theses and Dissertations (ETD). Paper 370. http://dx.doi.org/10.21007/etd.cghs.2013.0289.

This Dissertation is brought to you for free and open access by the College of Graduate Health Sciences at UTHSC Digital Commons. It has been accepted for inclusion in Theses and Dissertations (ETD) by an authorized administrator of UTHSC Digital Commons. For more information, please contact [email protected]. Cost of Illness Study of Anxiety Disorders for the Ambulatory Adult Population of the United States

Document Type Dissertation

Degree Name Doctor of Philosophy (PhD)

Program Health Outcomes and Policy Research

Research Advisor David K. Solomon, Pharm.D.

Committee James E. Bailey, M.D. Lawrence M. Brown, Pharm.D. Ph.D. Brandi E. Franklin, Ph.D. George Relyea, M.S.

DOI 10.21007/etd.cghs.2013.0289

Comments One year embargo expired in December 2014

This dissertation is available at UTHSC Digital Commons: https://dc.uthsc.edu/dissertations/370

Cost of Illness Study of Anxiety Disorders for the Ambulatory Adult Population of the United States

A Dissertation Presented for The Graduate Studies Council The University of Tennessee Health Science Center

In Partial Fulfillment Of the Requirements for the Degree Doctor of Philosophy From The University of Tennessee

By Elaheh Shirneshan December 2013

Copyright © 2013 by Elaheh Shirneshan. All rights reserved.

ii

DEDICATION

This dissertation is dedicated to my wonderful parents, Fatemeh and Masoud, to whom I owe every bit of my existence, to my beloved husband Ali, who has always believed in me and pushed me to do my best, and to my siblings, Emad and Afrooz, for their unconditional love and support.

iii

ACKNOWLEDGEMENTS

First, I would like to thank Dr. Lawrence M. Brown, and Dr. David K. Solomon, my research advisors, for all of their support and guidance. In addition, I would also like to thank my other committee members, Mr. Georg Relyea, Dr. Brandi E. Franklin, and Dr. Jim Bailey. Without their diligence and dedication, this dissertation would not have been possible.

iv

ABSTRACT

Background: Anxiety disorders are the most common psychiatric illness in the United States. However, economic burdens of this category of mental illnesses have not been well studied yet. The objective of this study was to estimate the societal cost of anxiety disorders for the ambulatory adult population of the U.S.

Data and Method: Data was collected from the 2009 and 2010 Medical Expenditure Panel Survey (MEPS), Bureau of Labor Statistics (BLS), and National Vital Statistics System (NVSS). Cost components included in the analyses were direct medical costs (i.e. cost for inpatient visits, outpatient visits, emergency room visits, office-based visits, prescription medications, and other services), and indirect cost (i.e. morbidity and mortality costs). Anxiety patients were identified using MEPS data. More specifically, individuals 18 years and older, who reported a diagnosis of, or had a medical event associated with anxiety disorder(s), were classified as anxiety population. Number of suicides due to anxiety disorders was estimated using the NVSS data. Direct medical costs attributable to anxiety disorders were estimated as the expenditures incurred by anxiety patients in excess of those incurred by anxiety-free population. Several multivariate regression analyses, using generalized linear models, were conducted to calculate the overall incremental direct medical costs associated with anxiety disorders, as well as cost by healthcare delivery setting, and cost for different sub-populations. Indirect costs were estimated using the Human Capital Approach (HCA). Morbidity cost was estimated by valuing the time period in which individuals had to stay in bed due to anxiety disorders. Mortality cost was estimated as the productivity loss from age at death to life expectancy.

Results: Among adult participants in 2009-2010 MEPS, 30.35 million (8.74%) reported being diagnosed with anxiety disorder(s). It was also estimated that in 2010, 3,497 suicides were due to anxiety disorders. The annual overall direct medical costs associated with anxiety disorders was estimated at $1657.52 per person (SE: $238.83; p <0.001), or $33.71 billion in total. Inpatient visits, prescription medications, and office- based visits together accounted for almost 93% of the overall cost. The increase in direct medical cost due to anxiety disorders was higher among White non-Hispanics ($1879.31) than Black non-Hispanics ($1459.30). For non-Hispanics, anxiety was not associated with a statistically significant increase in medical expenditure. Regarding aspects of indirect cost, morbidity and mortality cost were estimated at $12.72 billion and $2.34 billion in 2013 US dollars, respectively. The 2013 societal cost of anxiety disorders was estimated at $48.72 billion.

Conclusion: The current study demonstrates conclusively that anxiety disorders, with the annual cost of $48.72 billion in 2013 US dollars, absorb a significant portion of US healthcare resources and should be prioritized by policymakers and healthcare providers who aim to reduce downstream costs of mental disorders.

v

TABLE OF CONTENTS

CHAPTER 1. INTRODUCTION ...... 1 Importance of Conducting a Cost of Illness Study for Anxiety Disorders ...... 1 Why a Cost of Illness Analysis? ...... 1 Why Examining Costs by Service Category and for Different Sub-populations? ...... 2 Why Anxiety Disorders? ...... 2 The Existing Gap in the Literature ...... 3 Potential Contributions of This Research to the Current Literature ...... 4 Specific Aims ...... 6

CHAPTER 2. LITERATURE REVIEW ...... 7 An Overview of Anxiety Disorders ...... 7 Description of Anxiety Disorders ...... 7 Acute Stress Disorder ...... 7 Agoraphobia (with or without a History of Panic Disorder) ...... 8 Generalized Anxiety Disorder (GAD) ...... 8 Obsessive-compulsive Disorder (OCD)...... 9 Panic Disorder (with or without Agoraphobia) ...... 9 Phobias (Including Social Phobia) ...... 9 Post-traumatic Stress Disorder (PTSD) ...... 10 Epidemiology of Anxiety Disorders ...... 10 Treatment of Anxiety Disorders ...... 10 Medication ...... 11 Antidepressants ...... 11 Anti-anxiety drugs ...... 12 Betablockers ...... 12 Psychotherapy ...... 12 Diagnosis Specific Treatment ...... 13 An Overview of Cost of Illness Analysis ...... 14 Description of Cost of Illness Analysis...... 14 Direct and indirect costs...... 14 Perspective ...... 14 Incidence-based versus prevalence-based approach ...... 14 Measuring indirect costs ...... 15 Top-down or bottom-up approach ...... 15 History of Cost of Illness Studies ...... 15 Review of Cost of Illness Studies for Anxiety Disorders ...... 16 Studies in Which the Main Purpose Was to Calculate COI for Anxiety Disorders ..16 COI studies for multiple diagnoses of anxiety disorders ...... 16 COI studies for a particular diagnosis of anxiety disorders ...... 16 Studies in Which Obtaining the COI Was Not the Main Purpose of the Study ...... 17 Specific Features of the COI Studies for Anxiety Disorders ...... 17 Source of data ...... 17 Top-down or bottom-up approach ...... 17

vi

Perspective of the study ...... 19 Cost categories ...... 19 Incremental cost approach ...... 19

CHAPTER 3. METHODOLOGY ...... 21 Research Design ...... 21 Perspective ...... 21 Approach ...... 21 Data ...... 21 Sampling ...... 22 MEPS sampling plan...... 22 Sample size ...... 23 Study sample ...... 24 Data Overview ...... 24 MEPS Data Files ...... 24 Variables used in analyses from MEPS data files ...... 25 Summary variables...... 25 BLS Data ...... 31 NVSS Data ...... 31 Analysis Technique ...... 32 SA1: Estimating the Societal Cost of Anxiety Disorders for the U.S. Adult Population ...... 32 Overall direct medical cost...... 32 Morbidity cost ...... 35 Mortality cost ...... 37 SA2: Estimating the Incremental Direct Medical Expenditures Associated with Anxiety Disorders by Service Category...... 38 SA3: Estimating the Incremental Direct Medical Expenditures Associated with Anxiety Disorders for Different Sub-populations ...... 38 General Notes ...... 39

CHAPTER 4. RESULTS ...... 41 Descriptive Statistics ...... 41 Prevalence of Self-reported Anxiety Disorders in MEPS ...... 41 Characteristics of the Study Population ...... 41 Direct Medical Costs Attributable to Anxiety Disorders ...... 46 Preliminary Statistical Analyses ...... 46 Checking for multicollinearity ...... 46 Finding the distribution of cost data ...... 48 Overall Incremental Direct Medical Expenditure Attributable to Anxiety Disorders ...... 52 Incremental Direct Medical Expenditure by Health Delivery Setting (SA2) ...... 52 Incremental Direct Medical Expenditure for Different Sub-populations (SA3) ...... 52 Morbidity Cost Associated with Anxiety Disorders ...... 59 Number of Missed Work Days due to Anxiety Disorders for Employed Individuals ...... 59

vii

Checking for multicollinearity ...... 59 Finding the distribution of missed work days ...... 61 Number of Days Stayed in Bed due to Anxiety Disorders for Unemployed Individuals ...... 61 Daily Wage for Employed Individuals ...... 64 Average Daily Wage of Household Services for Unemployed Individuals ...... 64 Summary of Analyses ...... 64 Mortality Costs Associated with Anxiety Disorders ...... 64 Number of People Who Committed Suicide due to Anxiety Disorders and Age at Death ...... 66 Life Expectancy at Age of Death by Race/Ethnicity and Gender...... 66 Labor Force Participation Rate and Wage ...... 69 Summary of Analyses ...... 69 The Societal Cost of Anxiety Disorders for the U.S. Adult Population in 2010 (SA1) ...... 69

CHAPTER 5. DISCUSSION ...... 72 Direct Medical Costs ...... 72 General Notes ...... 72 Overall Direct Medical Cost ...... 73 Cost by Category of Healthcare Services ...... 73 Cost for Different Sub-populations ...... 74 Indirect Costs ...... 75 Societal Cost ...... 76 Limitations ...... 77

LIST OF REFERENCES ...... 79

VITA...... 87

viii

LIST OF TABLES

Table 2-1. Prevalence of anxiety disorders for the U.S. adult population...... 11

Table 2-2. Data sources used in COI studies of anxiety disorders...... 18

Table 2-3. Cost categories included in cost of illness studies...... 20

Table 3-1. Data sources for calculation of each cost component...... 22

Table 3-2. List of variables in MEPS data files used in the current study...... 26

Table 3-3. List of categorical variables...... 29

Table 3-4. Outcome variables and the initial set of covariates for estimating the incremental number of missed work days/ bed days due to anxiety disorders...... 36

Table 3-5. Sub-populations along with model specifications...... 40

Table 4-1. Comparison of demographic characteristics between adults with and without anxiety disorders...... 42

Table 4-2. Comparison of CCI clinical conditions between anxiety and non-anxiety patients...... 45

Table 4-3. Results of regression analysis to check for multicollinearity...... 47

Table 4-4. Results of the Breusch-Pagan/Cook-Weisberg test for heteroskedasticity. ..51

Table 4-5. Results of Park test...... 51

Table 4-6. Comparison of Poisson and Gamma variance functions...... 51

Table 4-7. Results of regression analysis to estimate the overall incremental direct medical expenditure associated with anxiety disorders...... 53

Table 4-8. Results of regression analyses to estimate the incremental expenditures of anxiety disorders by service category...... 54

Table 4-9. Incremental direct medical expenditures associated with anxiety disorders for different sub-populations...... 56

Table 4-10. Breaking down the cost of anxiety disorders among different race/ethnicities by gender, age, and geographic region...... 58

Table 4-11. Results of multicollinearity analysis for modeling the number of missed work days...... 60

ix

Table 4-12. Results of the Breusch-Pagan/Cook-Weisberg test for heteroskedasticity. ..63

Table 4-13. Results of Park test...... 63

Table 4-14. Incremental number of missed work days due to anxiety disorders...... 63

Table 4-15. Incremental number of bed days due to anxiety disorders...... 63

Table 4-16. Average daily wage for unemployed individuals based on BLS data...... 65

Table 4-17. Summary of results: Morbidity cost...... 65

Table 4-18. Number of suicides in 2010 by age, gender, and race/ethnicity...... 67

Table 4-19. Life expectancy at selected ages by race, Hispanic origin, race for non- Hispanic population, and sex: United States, 2010...... 68

Table 4-20. Labor force participation rate by age, gender, and race/ethnicity...... 70

Table 4-21. Median usual weekly earnings in current dollar from current population survey for 2010...... 70

Table 4-22. Mortality cost: Summary of results...... 71

Table 4-23. The societal cost of anxiety disorders for the U.S. adult population in 2010...... 71

x

LIST OF FIGURES

Figure 3-1. Overlapping design of MEPS sample...... 23

Figure 4-1. Distribution of the overall healthcare cost for the study population...... 49

Figure 4-2. Distribution of the log-transformed overall healthcare cost for the study population...... 50

Figure 4-3. Distribution of the number of missed work days for employed individuals...... 62

Figure 5-1. Percentage of each cost component from the societal cost...... 77

xi

LIST OF ABBREVIATIONS

ACA Affordable Care Act

AHRQ Agency for Healthcare Research and Quality

BLS Bureau of Labor Statistics

CBT Cognitive Behavioral Therapy

CC Clinical Classification

CCI Charlson Comorbidity Index

CI Confidence Interval

CMS Centers for Medicare & Medicaid Services

COI Cost of Illness

CPI Consumer Price Index

DSM IV-TR Diagnostic and Statistical Manual of Mental Disorders, fourth edition, text revision

GAD Generalized Anxiety Disorder

GDP Gross Domestic Product

GED Graduate Equivalency Degree

GLM Generalized Linear Model

HMO Health Maintenance Organization

ICD-9 International Classification of Diseases, Ninth Revision

LFPR Labor Force Participation Rate

MEPS Medical Expenditure Panel Survey

MEPS-HC Medical Expenditure Panel Survey -Household Component

MOI Monoamine Oxidase Inhibitor

xii

MSA Metropolitan Statistical Area

NCHS National Center for Health Statistics

NCS-R National Comorbidity Survey Replication

NIMH National Institute of Mental Health

NVSS National Vital Statistics System cost of illness (COI)

OCD Obsessive Compulsive Disorder

OECD Organization for Economic Cooperation and Development

OLS Ordinary Least Square

PHS Perceived Health Status

PTSD Post-traumatic Stress Disorder

SA Specific Aim

SE Standard Error

SSRI Selective Serotonin Reuptake Inhibitor

VIF Variance Inflation Factor

xiii

CHAPTER 1. INTRODUCTION

Data on the economic burden of anxiety disorders are limited. But this category of mental illnesses is known to be the most prevalent, and one of the most expensive psychiatric disorders in the U.S. The current study intends to answer the following research question: What are the economic burdens of anxiety disorders to the ambulatory adult population of the U.S.?

The following sections describe the importance of conducting a cost of illness study for anxiety disorders, the existing gap in the relevant literature, the potential contributions of this research to the current literature, and detailed explanation of specific aims of the study.

Importance of Conducting a Cost of Illness Study for Anxiety Disorders

Why a Cost of Illness Analysis?

According to Centers for Medicare & Medicaid Services (CMS) “U.S. health care spending reached $2.7 trillion in 2011, or $8,680 per person.”1(p. 2) There has been a growing rate of 3.9 percent in health spending since 2009. Also from 2009 to 2011, health spending was 17.9 percent of Gross Domestic Product (GDP). In comparison with other countries in the Organization for Economic Cooperation and Development (OECD), the United States has the highest health spending in terms of share of GDP. In fact, “Americans spent more than twice as much as relatively rich European countries such as France, Sweden and the United Kingdom.”2(p. 1) CMS has projected for this share to rise to 20 percent of GDP by 2020.

There is no simple and straightforward solution for controlling this high and ever- increasing rate of spending on healthcare in the United States; but, identifying the most expensive medical conditions and trying to alleviate their economic burden, through disease management interventions, is definitely a required step to be undertaken in this way. According to Joel & Segel, “Cost-of-illness studies measure the economic burden of a disease or diseases and estimate the maximum amount that could potentially be saved or gained if a disease were to be eradicated.”3(p. 2) In developing cost-containment policies and interventions, cost of illness (COI) analysis enables policymakers to identify diseases which need to be addressed with the highest priority.

No one can argue that healthcare resources, like many other forms of resource, are scarce; and the more resources absorbed by heath sector, the less will be left for other sectors such as education, defense, etc. A cost of illness study is not enough to set priorities for the objective of resource allocation (for that purpose, one needs effectiveness data as well as cost data). However, it is a main component of any cost- effectiveness or cost-utility analysis. Nowadays, cost of illness studies are being widely

1

used by health economists, pharmaceutical companies, clinicians, and policymakers to set research agendas and allocate resources.4,5

Why Examining Costs by Service Category and for Different Sub-populations?

Even though cost of illness analysis is a crucial primary step in identifying costly medical conditions, further information is needed if effectively reducing the burden of these conditions is desired. To reduce the burden of illness, just knowing the dollar amount in terms of cost of illness is not enough. In fact, one also needs to know the distribution of costs among different health sectors (inpatient, outpatient, emergency room, prescription medication …) and subpopulations (based on gender, race, age …) to find out if resources are being distributed disproportionally. Knowing which subpopulations or health sectors incur higher costs, policymakers can come up with tailored disease management interventions to address high costs specifically in those sub- populations/health sectors.

Healthcare costs are not distributed proportionally among different sub- populations. According to the Medical Expenditure Panel Survey (MEPS), in 2010, individuals with some college degree had the highest total healthcare expenditure in comparison with those in other educational levels. The same can be said for Whites, females, and married individuals. So having the estimation of cost of illness for the whole population (who suffer from that disease), one also needs to know how this burden is distributed among different groups of people. In the same manner, distribution of cost among different service categories will yield useful information as to which category of health services may benefit the most from disease management programs.

Why Anxiety Disorders?

Costly medical conditions can be amongst the underlying causes of skyrocketing healthcare expenditures in the U.S. In this regard, it is good to know that one group of these expensive medical conditions are anxiety disorders. Anxiety is a natural response and a necessary warning adaptation in humans. Everybody has experienced anxiety in the form of increased heart rate and tensed muscles. However, anxiety can become a pathologic disorder when it is excessive and uncontrollable; In other words, when it occurs without any recognizable reason or when the reason does not warrant such a reaction. If no medical condition accounts for symptoms, they are attributed to anxiety disorders.6

According to the Diagnostic and Statistical Manual of Mental Disorders, fourth edition, text revision (DSM IV-TR),7 anxiety disorders categorize a large number of disorders including acute stress disorder, post-traumatic stress disorder (PTSD), phobias (including social phobia), agoraphobia (with or without a history of panic disorder), panic disorder (with or without agoraphobia), generalized anxiety disorder (GAD), obsessive- compulsive disorder (OCD), and anxiety disorders due to known physical causes (these

2

include general medical conditions or substance abuse). The primary feature of these disorders is abnormal or inappropriate anxiety. These disorders usually interfere with a patient’s work, schooling, and family life so that in severe cases, they can adversely affect their function. For instance, anxiety disorders can be an underlying cause of alcohol and substance abuse.

Fortunately, anxiety disorders are highly treatable, and most people with these conditions can have fulfilling lives if they receive appropriate treatment. In general, anxiety disorders are treated with medication, specific types of psychotherapy, or both. “Treatment choices depend on the problem and the person’s preference. Before treatment begins, a doctor must conduct a careful diagnostic evaluation to determine whether a person’s symptoms are caused by an anxiety disorder or a physical problem.”8(p. 14)

Each year, almost one out of three Americans age 18 years or older (12-month prevalence=26.2%), will experience at least one form of mental disorders (i.e. anxiety disorders, mood disorders, impulse-control disorders, and substance use disorders).9 More than two thirds of these individuals (12-month prevalence=18.1%) suffer from one (or more) form of anxiety disorders (alone or along with another mental condition).9 In other words, anxiety disorders represent the most common psychiatric illnesses in the U.S. The annual societal cost of anxiety disorders in the U.S. in 1990 was estimated to range from US$42 to US$47 billion, nearly approaching the estimates of the cost of depression (from US$44 to US$53 billion) for the same time frame.5,10 With respect to the high prevalence of anxiety disorders in the U.S. and evidence from previous research,5,10 it is expected that this category of mental illnesses imposes a significant economic burden to society. In this regard, a more current and comprehensive evaluation of healthcare expenditures attributable to anxiety disorders is warranted. Such evaluation may guide clinicians and policymakers in developing and improving disease management programs, and provide a basis for conducting cost-effectiveness analysis of new treatment interventions.

The Existing Gap in the Literature

In order to have a reliable estimation of the economic burden of a disease to society, there are several factors that must be taken into account:

x Both direct and indirect costs should be included in the analysis.

x The sample should be representative of the entire population in which results are to be generalized to.

x If there is more than one diagnosis for a condition, all possible diagnoses should be considered.

x In estimating the costs, not only the costs directly due to the condition of interest should be included, but also costs due to the comorbidities and complications resulted from that condition should be considered.

3

x When using different data sources, the potential inconsistency between these data sources should be taken into account.

An overview of the literature on cost of illness studies for anxiety disorders reveals that there are a limited number of studies in this field. In fact, the only studies that estimated direct and indirect costs of anxiety disorders from a societal perspective in the U.S. were conducted far back, in 1996 and 1999 by DuPont et al.5 and Greenberg et al.10

DuPont et al.5 conducted a cost of illness analysis from a societal perspective, by including direct medical costs, direct non-medical costs, and indirect costs (morbidity and mortality costs). The major limitation of their study is that in estimating the cost of illness for anxiety disorders, they only considered the costs that were incurred directly due to anxiety disorders; but they did not account for the increase in general use of medical services in patients suffering from anxiety disorders. In addition, they did not include all diagnoses of anxiety disorders in their analysis (data on costs due to PTSD and acute stress disorder were not collected). All of this might have led to an underestimation of the true burden of anxiety disorders. Another limitation of the study is related to the data collection process. Data on utilization and cost was collected from several different data sources. For instance, they used more than six databases (such as National Hospital Discharge Survey, National Nursing Home Survey, Pharmaceutical Data Source …) for calculating direct medical costs. The potential inconsistencies between different data sources may later cast doubt on the reliability of results.

Greenberg et al.10 reduced the inconsistency in data by using less data sources. They collected the majority of data from the National Comorbidity Survey. The cost components included in their study were direct psychiatric costs, direct non-psychiatric costs, and indirect costs (morbidity and mortality costs). Direct non-psychiatric costs accounted for most of the cost of illness for anxiety disorders. However, estimation of this cost component (which accounted for more than half of their estimated cost of illness) was based on results from a single staff–model HMO that may not be fully generalizable to the entire population. So, their estimation of cost of illness for anxiety disorders may not be representative of the true burden of this condition to the whole society. In addition, they did not include all diagnoses of anxiety disorders in their analysis (data on costs due to OCD and acute stress disorder were not collected).

In more recent studies, the study population is either not from the U.S. or not representative of the whole U.S. population,11-15 the perspective is not societal,11,16-22 both direct and indirect costs are not estimated,11,15-23 or not all anxiety diagnoses are included in the study.16,17,24-29

Potential Contributions of This Research to the Current Literature

Due to the high prevalence of anxiety disorders in the U.S. and the evidence from previous research on anxiety-related costs,5,10 it is expected that this category of mental illnesses imposes a significant economic burden to society. Considering the fact that

4

costly medical conditions have been always a high priority to policymakers in their effort to reduce healthcare costs, a well-conducted cost of illness for anxiety disorders can provide them with a reliable estimation of the economic burden of these conditions. Moreover, identifying the distribution of costs among different sub-populations and health sectors may enable them to come up with tailored disease management programs to reduce the costs.

The specific features of the study which distinguish it from the previous works, and also add to the current knowledge are as follows:

x Results of this research will provide the most comprehensive and updated estimate of cost of illness for anxiety disorders, for the ambulatory adult population of the U.S.

x It is the first study that measures costs at the population level, as well as for different sub-populations.

x The sample is representative of the whole U.S civilian non-institutionalized population 18 years of age and older. So, results can be generalized to this group of individuals (Civilian-non-institutionalized population refers to community- dwelling population or ambulatory population. individuals in the military and those residing in nursing homes, assisted living facilities, and prisons are not included in the civilian-non-institutionalized population. Throughout this research, civilian non-institutionalized population and ambulatory population phrases are used alternatively).

x The societal perspective enables the researcher to measure the economic burdens of anxiety disorders to the whole society regardless of who incurred the costs.

x Instead of getting data from several different data sources, which may lead to inconsistency in data and later affect the reliability of results, only one database will be used to collect data on healthcare utilization and costs.

x All possible diagnoses of anxiety disorders will be included in the analysis.

x The estimation of cost of illness will not only capture the costs directly due to anxiety disorders, but it will also include cost of their comorbidities and complications.

x The analysis technique used will control for all factors that may affect healthcare expenditures, therefore, researchers can estimate costs solely due to anxiety disorders.

5

Specific Aims

Specific aims of the study are presented in this section. For all specific aims, estimates will be provided for the ambulatory adult (18 years and older) population of the U.S.

x Specific Aim 1 (SA1): To estimate direct medical cost and indirect costs (morbidity cost, mortality cost) of anxiety disorders.

x Specific Aim 2 (SA2): To estimate direct medical costs attributable to anxiety disorders by major service categories (i.e. inpatient visits, outpatient visits, office- based medical visits, emergency room visits, prescription medications, and other services).

x Specific Aim 3 (SA3): To estimate direct medical costs attributable to anxiety disorders for different sub-populations (based on gender, race/ethnicity, age, marital status, poverty category, education, geographic region, Metropolitan Statistical Area (MSA), and insurance coverage).

6

CHAPTER 2. LITERATURE REVIEW

The goal of this chapter is to provide a review of literature relevant to the purpose of this research. It has two main sections. The first section provides an introduction to anxiety disorders, their epidemiology and treatments. The second section is devoted to cost of illness studies. It starts with description and history of cost of illness studies and continues with reviewing the cost of illness studies for anxiety disorders. In order to find these studies, we conducted a search in Pubmed, Ovid, PsycINFO, and Google Scholar for articles published after 1990. The search words were cost, expenditure, cost of illness, burden, and anxiety disorders.

An Overview of Anxiety Disorders

Description of Anxiety Disorders

According to the American Psychiatric Association,7 anxiety disorders categorize a large number of disorders including acute stress disorder, agoraphobia (with or without a history of panic disorder), generalized anxiety disorder (GAD), obsessive-compulsive disorder (OCD), panic disorder (with or without Agoraphobia), phobias (including social phobia), and post-traumatic stress disorder (PTSD). The primary feature of these disorders is abnormal or inappropriate anxiety. Everybody has experienced anxiety in forms of increased heart rate and tensed muscles. But anxiety becomes a problem when it occurs without any recognizable reason, or when the reason does not warrant such a reaction. If no medical condition accounts for symptoms, they are attributed to anxiety disorders.6

The following sections provide information on etiology, symptoms and prognosis of different diagnoses of anxiety disorders, quoted from AllPsych Online (http://allpsych.com/disorders/anxiety/index.html).6

Acute Stress Disorder

x Etiology: “By definition, acute stress disorder is a result of a traumatic event in which the person experienced or witnessed an event that involved threatened or actual serious injury or death and responded with intense fear and helplessness.”6

x Symptoms: “Symptoms include dissociative symptoms such as numbing, detachment, a reduction in awareness of the surroundings, derealization, or depersonalization; re-experiencing of the trauma, avoidance of associated stimuli, and significant anxiety, including irritability, poor concentration, difficulty sleeping, and restlessness. The symptoms must be present for a minimum of two days and a maximum of four weeks and must occur within four weeks of the

7

traumatic event for a diagnosis to be made.”6

x Prognosis: “Prognosis for this disorder is very good. If it should progress into another disorder, success rates can vary according to the specific of that disorder.”6

Agoraphobia (with or without a History of Panic Disorder)

x Etiology: “Agoraphobia can develop out of simple phobias or it can be a result of extreme trauma, although it is often a result of numerous panic attacks such as those found in panic disorder.”6

x Symptoms: “Agoraphobia, like other phobias, is made up of extreme anxiety and fear. Different from other phobias, however, is the generalization which occurs. Agoraphobia is the anxiety about being in places where escape might be difficult or embarrassing or in which help may not be available should a panic attack develop. It can be sub diagnosed as either ‘with’ or ‘without’ panic disorder. Typically situations that invoke anxiety are avoided and in extreme cases, the person may never or rarely leave their home.”6

x Prognosis: “Prognosis is good, especially if the individual has some insight into the development of the disorder and if their fears are irrational and there is insight into this.”6

Generalized Anxiety Disorder (GAD)

x Etiology: “Often anxiety gets generalized to other situations, and can then become overwhelming or associated with life in general. Typically GAD develops over a period of time and may not be noticed until it is significant enough to cause problems with functioning.”6

x Symptoms: “As its name implies, GAD is evidenced by general feelings of anxiety such as mild heart palpitations, dizziness, and excessive worry. The symptoms are difficult to control for the individual and are not related to a specific event (such as in PTSD) and are not as severe as those found with Panic Disorder.”6

x Prognosis: “Prognosis is good for the more extreme symptoms, but those associated with underlying fears are more difficult to treat (such as excessive worry). Working through childhood issues can be helpful as these tend to get distorted as they follow us into adulthood (e.g., over-controlling parental styles, sexual abuse, and childhood phobias).”6

8

Obsessive-compulsive Disorder (OCD)

x Etiology: “Both biological and psychological causes have been found in OCD.”6

x Symptoms: “The key features of this disorder include obsessions (persistent, often irrational, and seemingly uncontrollable thoughts) and compulsions (actions which are used to neutralize the obsessions). A good example of this would be an individual who has thoughts that he is dirty, infected, or otherwise unclean which are persistent and uncontrollable. In order to feel better, he washes his hands numerous times throughout the day, gaining temporary relief from the thoughts each time. For these behaviors to constitute OCD, it must be disruptive to everyday functioning (such as compulsive checking before leaving the house making you extremely late for all or most appointments, washing to the point of excessive irritation of your skin, or inability to perform everyday functions like work or school because of the obsessions or compulsions).”6

x Prognosis: “Prognosis for this disorder has a wide range, depending upon how the individual responds to medication and how deep rooted the underlying issues are.”6

Panic Disorder (with or without Agoraphobia)

x Etiology: “Often the symptoms of this disorder come on rapidly and without an identifiable stressor. The individual may have had periods of high anxiety in the past, or may have been involved in a recent stressful situation. The underlying causes, however, are typically subtle.”6

x Symptoms: “Panic Disorder is characterized by sudden attacks of intense fear or anxiety, usually associated with numerous physical symptoms such as heart palpitations, rapid breathing or shortness of breath, blurred vision, dizziness, and racing thoughts. Often these symptoms are thought to be a heart attack by the individual, and many cases are diagnosed in hospital emergency rooms.”6

x Prognosis: “Prognosis for this disorder is very good if the above conditions are met. Left untreated, however, symptoms can worsen and Agoraphobia can develop. In these cases, the individual has developed such an intense fear that leaving the safety of home feels impossible.”6

Phobias (Including Social Phobia)

x Etiology: “Often a traumatic event is the precursor for a phobia, which may or may not be at the conscious level.”6

x Symptoms: “Symptoms include either extreme anxiety and fear associated with

9

the object or situation or avoidance. To be diagnosed, the symptoms must be disruptive to everyday functioning (such as quitting a great job merely because you have to use an elevator).”6

x Prognosis: “Prognosis is very good if treated effectively.”6

Post-traumatic Stress Disorder (PTSD)

x Etiology: “By definition, PTSD always follows a traumatic event which causes intense fear and/or helplessness in an individual. Typically the symptoms develop shortly after the event, but may take years. The duration for symptoms is at least one month for this diagnosis.”6

x Symptoms: “Symptoms include re-experiencing the trauma through nightmares, obsessive thoughts, and flashbacks (feeling as if you are actually in the traumatic situation again). There is an avoidance component as well, where the individual avoids situations, people, and/or objects which remind him or her about the traumatic event (e.g., a person experiencing PTSD after a serious car accident might avoid driving or being a passenger in a car). Finally, there is increased anxiety in general, possibly with a heightened startle response (e.g., very jumpy, startle easy by noises).”6

x Prognosis: “Prognosis ranges from moderate to very good. Those with the best prognosis include situations where the traumatic event was acute or occurred only one time (e.g., car accident) rather than chronic or on-going trauma (e.g., ongoing sexual abuse, war).”6

Epidemiology of Anxiety Disorders

For any anxiety disorder, 12-month prevalence is 18.1% of the U.S. adult population.9 Lifetime prevalence is 28.8% of the U.S. adult population and 25.1% of children and adolescents.30 Women are 60% more likely than men to experience an anxiety disorder over their lifetime. Non-Hispanic Blacks are 20% less likely, and Hispanics are 30% less likely, than non-Hispanic Whites to experience an anxiety disorder during their lifetime. Individuals in the age group of 30-44 have the highest risk of developing an anxiety disorder over their lifetime (35.1%), while those who are 60 or older have the lowest risk (15.3%).30 Table 2-1 shows the epidemiology of anxiety disorders in the U.S. adult population for each specific diagnosis.

Treatment of Anxiety Disorders

According to the National Institute of Mental Health (NIMH) “anxiety disorders are treated with medication, specific types of psychotherapy, or both. Treatment choices

10

Table 2-1. Prevalence of anxiety disorders for the U.S. adult population.

Diagnosis Lifetime prevalence 12-month prevalence Any Anxiety Disorder 28.8% 18.1% Acute Stress Disorder 7.8% - Agoraphobia 1.4% 0.8% Generalized Anxiety Disorder 5.7% 3.1% Social Phobias 12.1% 6.8% Specific Phobias 12.5% 8.7% Post-traumatic Stress Disorder 6.8% 3.5% Obsessive-compulsive Disorder 1.6% 1.0% Panic Disorder 4.7% 2.7%

Source: Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. Jun 2005;62(6):617-627; Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. Jun 2005;62(6):593-602.

depend on the problem and the person’s preference. Before treatment begins, a doctor must conduct a careful diagnostic evaluation to determine whether a person’s symptoms are caused by an anxiety disorder or a physical problem.”8(p. 14)

The following sections, quoted from the Booklet of Anxiety Disorders,8 describe treatment options for anxiety disorders.

Medication

“Medication will not cure anxiety disorders, but it can keep them under control while the person receives psychotherapy.”8(p. 14) Antidepressants, anti-anxiety drugs, and betablockers are the three major categories medications used for anxiety disorders.

Antidepressants. Even though Antidepressants were originally developed to treat depression, they are also effective in treating anxiety disorders.8 The specific antidepressant used to treat anxiety disorders are listed below:

x Selective serotonin reuptake inhibitors (SSRIs): “SSRIs alter the levels of the neurotransmitter serotonin in the brain, which, like other neurotransmitters, helps brain cells communicate with one another. Fluoxetine (Prozac®), sertraline (Zoloft®), escitalopram (Lexapro®), paroxetine (Paxil®), and citalopram (Celexa®) are some of the SSRIs commonly prescribed for panic disorder, OCD, PTSD, and social phobia. SSRIs are also used to treat panic disorder when it

11

occurs in combination with OCD, social phobia, or depression. Venlafaxine (Effexor®), a drug closely related to the SSRIs, is used to treat GAD. These medications are started at low doses and gradually increased until they have a beneficial effect. SSRIs have fewer side effects than older antidepressants. Their common side effects are slight nausea or jitters (when people first start to take them) as well as sexual dysfunction.”8(p. 15)

x Tricyclics: “Tricyclics are older than SSRIs and work as well as SSRIs for anxiety disorders other than OCD. Tricyclics include imipramine (Tofranil®), which is prescribed for panic disorder and GAD, and clomipramine (Anafranil®), which is the only tricyclic antidepressant useful for treating OCD. They are also started at low doses that are gradually increased. They sometimes cause dizziness, drowsiness, dry mouth, and weight gain.”8(p. 15)

x MAOIs: “Monoamine oxidase inhibitors (MAOIs) are the oldest class of antidepressant medications. The MAOIs most commonly prescribed for anxiety disorders are phenelzine (Nardil®), followed by tranylcypromine (Parnate®), and isocarboxazid (Marplan®),which are useful in treating panic disorder and social phobia. If taken with food and beverages containing tyramine or certain medications, including some types of birth control pills, pain relievers, cold and allergy medications, and herbal supplements drug interaction will occur which leads to increase in blood pressure.”8(p. 16)

Anti-anxiety drugs. “High potency benzodiazepines combat anxiety and its main side effect is that patient may get dependent to it. Clonazepam (Klonopin®) is used for social phobia and GAD, lorazepam (Ativan®) is helpful for panic disorder, and alprazolam (Xanax®) is useful for both panic disorder and GAD. This drug in now less prescribed due to its side effects.”8(p. 16)

Betablockers. “Betablockers such as propranolol (Inderal®), which is used to treat heart conditions, can prevent the physical symptoms that accompany certain anxiety disorders, particularly social phobia.”8(p. 17)

Psychotherapy

“Psychotherapy involves talking with a trained mental health professional, such as a psychiatrist, psychologist, social worker, or counselor, to discover what caused an anxiety disorder and how to deal with its symptoms.”8(p. 17) One of the specific forms of psychotherapy, which is very effective in treating some forms of anxiety disorders, is called Cognitive Behavioral Therapy (CBT). According to the booklet of NIMH on anxiety disorders:

CBT is very useful in treating anxiety disorders. The cognitive part helps people change the thinking patterns that support their fears, and the behavioral part helps people change the way they react to anxiety-provoking situations. There is some

12

evidence that the benefits of CBT last longer than those of medication for people with panic disorder, and the same may be true for OCD, PTSD, and social phobia. If a disorder recurs at a later date, the same therapy can be used to treat it successfully a second time. Medication can be combined with psychotherapy for specific anxiety disorders, and this is the best treatment approach for many people.8(p. 17)

Diagnosis Specific Treatment

The following sections, quoted from AllPsych Online (http://allpsych.com/disorders/anxiety/index.html),6 provide treatment recommendations for each specific diagnosis of anxiety disorder.

x Acute Stress Disorder: “The disorder may resolve itself with time or may develop into a more severe disorder such as PTSD. Medication can be used for a very short duration (up to four weeks) or psychotherapy can be utilized to assist the victim in dealing with the fear and sense of helplessness.”6

x Agoraphobia: “(with or without a history of panic disorder): Treatment may involve anxiety reduction techniques aimed at increasing the control a person feels over his or her anxiety and fears. Other approaches require the individual to work through their anxiety in relation to interpersonal or childhood issues.”6

x Generalized Anxiety Disorder (GAD): “Medication and/or psychotherapy have been found to be helpful, especially therapy aimed at teaching the client how to gain control over the symptoms.”6

x Obsessive-compulsive Disorder (OCD): “Medication is often prescribed for individuals with OCD. Psychotherapy can be helpful in learning ways to feel more in control, cope better with stressors, and explore the underlying issues associated with the obsessive thoughts.”6

x Panic Disorder (with or without Agoraphobia): “Although medication can be useful, psychotherapy (especially behavioral and cognitive/behavioral approaches have proved quite successful). The key to treatment is accepting the panic attacks as psychological rather than physical (once these causes have been ruled out by a physician), practicing relaxation exercises, and working through the underlying issues.”6

x Phobias (including Social Phobia): “Treatment is often behavioral in nature, with the therapist guiding the client through exercises more closely resembling the feared object or situation. Exploring underlying issues can also be beneficial.”6

x Post-traumatic Stress Disorder (PTSD): “Psychological treatment is considered the most effective means to recovery from PTSD, although some medications

13

(such as anti-anxiety meds) can help alleviate some symptoms during the treatment process.”6

An Overview of Cost of Illness Analysis

Description of Cost of Illness Analysis

According to Joel & Segel, “Cost-of-illness studies measure the economic burden of a disease or diseases and estimate the maximum amount that could potentially be saved or gained if a disease were to be eradicated.”3(p. 2) Both direct and indirect costs are included in a comprehensive analysis. However, based on the perspective of a study, either of these cost categories can be excluded.3

Direct and indirect costs. Direct costs refer to the opportunity cost of resources used for treating a particular disease. However in measuring indirect costs, the main focus is on the value of resources lost due to a particular disease.31 According to Hodgson and Meiners, opportunity cost is defined as “the value of the forgone opportunity to use in a different way those resources that are used or lost due to illness.”3(p. 4)

Direct costs include direct medical costs and nonmedical direct costs. Direct medical costs include costs for hospital inpatient stays, emergency room visits, office- based medical visits, outpatient visits, prescription medication, nursing home care, hospice care, rehabilitation care, home health care, and cost for medical supplies.31,32 Nonmedical direct costs include costs due to transportation to health care facilities; relocation and any other change in one’s life pattern due to the illness.3 Indirect costs include mortality cost (cost of premature death due to the illness) and morbidity cost (productivity loss).

There are studies in which intangible costs i.e. costs of pain and suffering are included in the analysis as well. However, estimation of intangible costs is less common due to difficulty in measuring these costs.3

Perspective. One of the most important aspects of any economic evaluation is the perspective of the study. In fact, inclusion and exclusion of the above discussed cost categories depend on the perspective undertaken in the study. There are several different perspectives that can be used in economic evaluations. For instance, one perspective measures the costs to the society (societal perspective) and another one may take into account costs to the third party payer (payer perspective). There are other perspectives in which costs to businesses, the government, and participants and their families are included.4,31

Incidence-based versus prevalence-based approach. In an incidence-based study, the attempt is to estimate the lifetime cost for a patient with a particular illness. Here the lifetime cost refers to the cost of an illness from diagnosis to cure or death. On

14

the other hand, in prevalence-based studies, all costs for a particular population in a given time period (regardless of the date of onset) will be measured. Requiring less data and fewer assumptions than incidence-based studies, prevalence based studies are far more common.3

Measuring indirect costs. There are three primary approaches to estimate indirect costs including the Human Capital approach (HCA), the friction cost method, and the willingness to pay approach. In the HCA, the lost production for employed patients/caregivers is measured in terms of lost earnings.33,34 If the patient/care giver is not employed, the value of household work would be equivalent to the cost of hiring a replacement from the labor market.3

Friction cost method is a more realistic approach to measure indirect costs. The assumption here is that as long as unemployment rate is greater than zero, the market would be able to replace the lost work force. So, the loss of an employee will lead to production loss until the new employee is hired and trained.35-37 This period is called the friction period.

Finally, indirect costs can be measured using the willingness to pay approach. According to Joel & Segel, “The willingness to pay approach measures the amount an individual would pay to reduce the probability of illness or mortality.”3(p. 14)

Top-down or bottom-up approach. In top-down approach, highly aggregated data sources (on population-level) would be directly used. While in bottom-up approach, the individual-level data (adopted from patients, patient’s chart, epidemiological registries or cohort studies) would be aggregated to get the population-level data.38

History of Cost of Illness Studies

Cost-of-illness studies were among the first economic evaluation studies and first appeared in the literature in 1913 in a paper called The value of human life.39 Later in the 1950s and 1960s, other researchers tried to develop the cost of illness methodology further.34,40,41

In 1959, Mushkin and Collings tried to clarify cost concepts in cost of illness studies. They introduced a classification of costs “based on their effects on the use, distribution, and quantity of economic resources”.40(p. 795) In 1966, Dorothy Rice developed a framework for calculating single-year costs of illness, disability, and death by major category of illness, using existing data sets. She also tried to address the problems in which a researcher encounters in measuring direct and indirect costs.34 Later, this method became a standard approach for conducting a cost of illness study. Since 1966, Rice has updated and refined her methodology.

Cost of illness studies became more popular in the 1970s and 1980s, when they were used to gain support for more resources being devoted to health care.42 Later in

15

1990s, pharmaceutical companies started to use cost of illness studies to highlight the economic burden of a particular disease. The aim was to justify the fact that more resources, i.e. the company’s product, should be devoted to that particular disease.42 Nowadays, cost of illness studies are being widely used by health economists, pharmaceutical companies, clinicians, and policymakers to set research agendas and allocate resources.4,5

Review of Cost of Illness Studies for Anxiety Disorders

An overview of the literature on cost of illness studies for anxiety disorders revealed that there are quite a few studies in this field. In general, these studies form up two separate categories. One category includes the studies in which the main purpose was to calculate the cost of illness for anxiety disorders. This category by itself has two different subcategories: cost of illness studies for multiple diagnoses of anxiety disorders, and cost of illness studies for a particular diagnosis of anxiety disorder. Another major category consists of studies in which obtaining the cost of illness was not the main purpose of the study. For instance, some have calculated the cost as part of a cost- effectiveness study for a particular treatment of anxiety disorders, while some tried to determine the predictors of cost for anxiety disorders.

Studies in Which the Main Purpose Was to Calculate COI for Anxiety Disorders

In this section, studies with the objective of calculating cost of illness for anxiety disorders are reviewed.

COI studies for multiple diagnoses of anxiety disorders. DuPont et al.5 and Greenberg et al.10 estimated the direct and indirect costs of anxiety disorders in U.S. in 1990s. Goetzel et al.11 assessed the health and productivity cost burden of the top ten physical and mental health conditions, including anxiety disorders, affecting six large U.S. employers; while Marciniak et al.15 tried to estimate the cost of anxiety disorders among the employed individuals in the United States. Andlin-Sobocki et al.13 conducted a study to estimate the cost of anxiety disorders in Europe and Smit et al.14 estimated the costs of nine common mental disorders, including anxiety disorders, in Netherlands.

COI studies for a particular diagnosis of anxiety disorders. Siegel et al.,24 Leon et al.,16 and Batelaan et al.28 assessed the costs of panic disorder in the U.S., and Netherlands, respectively. While Patel et al.27 and Acarturk et al.29 estimated the economic consequences of social phobia in the Great Britain and Netherlands. Rees et al.17 compared medical utilization and costs incurred by people with panic disorder to those incurred by people with social phobia.

16

Studies in Which Obtaining the COI Was Not the Main Purpose of the Study

Below is the list of studies falling in this category:

x Salvador-Carulla et al.26 estimated the costs before and after the diagnosis and the provision of effective treatment for panic disorder.

x Souetre et al.25 examined how co-morbidity and symptom severity related to the costs of generalized anxiety disorder (GAD).

x Marciniak et al.18 examined the different clinical and demographic characteristics that could affect the cost of treating patients with anxiety disorders.

x Roberge et al.23 examined healthcare services utilization and costs before and after providing an empirically supported cognitive-behavioral treatment for panic disorder with agoraphobia.

x McLaughlin et al.20 conducted a study aimed at measuring the impact of having both depression and anxiety, having neither or either condition alone on treatment patterns, health care utilization, and cost.

x Stein et al.21 tried to find out how antidepressant adherence, among patients with anxiety disorders, will affect medical resource use and costs.

x Olfson and Gameroff 22 conducted a study to evaluate the extent to which pain severity contributes to the health care costs of patients with generalized anxiety disorder (GAD).

Specific Features of the COI Studies for Anxiety Disorders

In the following sections, each of the above-mentioned studies is assessed with respect to the main features of a cost of illness analysis.

Source of data. In order to get data on resource use and cost, almost half of the studies used retrospective cohort study methodology. In some of these studies, patients were asked retrospectively about their utilization,16,17,24,25,29 while in other studies, medical databases providing individual information on healthcare utilization and cost were used to obtain required data.5,10,11,13,15,18,20,21 Table 2-2 shows the list of these databases.

Top-down or bottom-up approach. Only two studies used the top-down approach to calculate cost of illness for anxiety disorders.5,10 Other studies used the bottom-up approach.

17

Table 2-2. Data sources used in COI studies of anxiety disorders.

Authors Data source DuPont et al.5 National Institute of Mental Health (NIMH) National Hospital Discharge Survey American Medical Association National Center for Health Statistics National Nursing Home Survey Pharmaceutical Data Source

Greenberg et al.10 National Comorbidity Study (NCS) A large staff–model health maintenance organization The U.S. Bureau of the Census Professional associations and news periodicals National Center for Health Statistics Industry sources

Goetzel et al.11 Health and Productivity Management (HPM)a Subset of MedStat’s MarketScanb Database

Andlin-Sobocki13 German National Health Interview and Examination Survey - mental health supplement (GHS–MHS)c

Marciniak et al.15 Health and Productivity Management (HPM) Subset of MedStat’s MarketScan Database

McLaughlin et al.20 PharMetrics Patient-Centric Databased

Stein et al.21 Integrated Healthcare Information Services National Managed Care Benchmark Databasee a The HPM database contains workplace absence, short-term disability, and worker compensation data for six fortune 200 U.S. employers. b The Market- Scan database is an annual medical database that includes private sector health data from approximately 100 payers and contains data on clinical utilization, expenditures, and enrollment across inpatient, outpatient, prescription drug, and carve-out services. This database links paid claims and encounter data to detailed patient information across sites and types of providers over time. c The survey was carried out in 1998/99 and included a community sample of 4181 (age: 18–65) individuals. d PharMetrics Patient-Centric database is composed of medical and pharmaceutical claims for approximately 36 million patients from 61 health plans across the United States. e Located in Waltham, Massachusetts, this nationally representative database includes data from 30 health plans covering more than 25 million persons.

18

Perspective of the study. Eight out of twenty one studies used payer perspective,11,15-17,19-22 while other studies used societal perspective.

Cost categories. More than half of the studies included indirect costs in their analysis as well as direct costs. To estimate indirect costs, all studies used HCA. Only four studies gave an estimation of indirect costs on a population level (employed or unemployed patients).5,13,29,43 The rest reported indirect costs based on employed patients only.10,11,14,15,24-28 Specific cost categories used in these studies are shown in Table 2-3.44 As one can see, the most frequent cost categories included in the analysis were outpatient treatment cost, costs for hospital treatment, drugs, and emergency room facilities. Studies conducted by DuPont et al.5 and Greenberg et al.10 were the most comprehensive studies with respect to cost categories included in the analysis.

Incremental cost approach. In estimating the cost of illness for anxiety disorders, less than half of the studies calculated the incremental healthcare cost of individuals with anxiety disorders, in excess of those who were anxiety free. In other words, not only costs directly due to anxiety disorders were included, but also costs due to comorbidities and complications of anxiety disorders were considered in their cost estimation;14,15,17,18,20,22,24,27,29 Rest of the studies only estimated the costs which were directly due to anxiety disorders.

19

Table 2-3. Cost categories included in cost of illness studies.

Direct costs Indirect costs Outpatient Inpatient Medications Rehabilitations ER Non Productivity Sick Early Mortality

-medical -medical -leave

retirement retirement

l Authors oss

Edlund and Sawn43 9 9 Siegel et al.24 9 9 9 Souetre et al.25 9 9 9 9 9 Salvador-Carulla et al.26 9 9 9 9 DuPont et al.5 9 9 9 9 9 9 9 9 9 9 Leon et al.16 9 Rees et al.17 9 9 Greenberg et al.10 9 9 9 9 9 9 9 9 9 Patel et al.27 9 9 9 9 Goetzel et al.11 9 9 9 9 9 Marciniak et al.15 9 9 9 9 Marciniak et al.18 9 9 9 Andlin-Sobocki et al.13 9 9 9 9 9 9 Panzer et al.19 9 9 9 9 Roberge et al.23 9 9 McLaughlin et al.20 9 9 9 9 Smit et al.14 9 9 9 9 9 9 9 Stein et al.21 9 9 9 9 Batelaan et al.28 9 9 9 9 9 9 9 Olfson and Gameroff 22 9 9 9 Acarturk et al.29 9 9 9 9 9

ER = Emergency Room. Source: Konnopka A, Leichsenring F, Leibing E, Konig HH. Cost-of-illness studies and cost-effectiveness analyses in anxiety disorders: a systematic review. Journal of Affective Disorders. Apr 2009; 114(1-3):14-31.

20

CHAPTER 3. METHODOLOGY

Research Design

In order to estimate the cost of illness for anxiety disorders, a retrospective database analysis was conducted. The specific features of the research are as follows:

Perspective

Cost of illness for anxiety disorders was calculated from a societal perspective. In other words, this economic evaluation includes the impact of anxiety disorders on the welfare of the whole society. In order to achieve this goal, both direct and indirect costs attributable to anxiety disorders have been estimated. The costs components included in this study are as follow:

x Direct medical costs: This category refers to expenditures for inpatient visits, outpatient visits, office-based medical provider visits, emergency room visits, prescription medications, and other medical expenses (other medical expenses refer to expenses not included in the above-mentioned categories, such as expenditures for home health and medical devices and supplies).

x Indirect costs: This category includes morbidity and mortality costs.

Approach

To estimate cost of illness, one can take either incidence-based or prevalence- based approach. As it was mentioned in the previous chapter, prevalence-based approach is far more common, since it needs less data and time in comparison with the incidence- based one. So in this research, a prevalence-based approach was adopted to estimate the cost of illness for anxiety disorders.

Data

Data was collected from three major databases. These databases are explained in details in the “Data Overview” section.

x Medical Expenditures Panel Survey (MEPS).

x The Bureau of Labor Statistics (BLS).

x The National Vital Statistics System (NVSS).

21

Table 3-1 shows the exact databases used for calculation of each cost component. Using these databases, the researcher estimated costs at individual level. The bottom-up approach was undertaken to aggregate individual-level data to population-level.

Sampling

MEPS sampling plan. The study sample was based on the MEPS sampling plan. As it was mentioned in chapter 2, MEPS collects data on health services utilization and cost, as well as how frequently these services are used and how they are paid.45

MEPS currently has two major components: the Household Component and the Insurance Component. The Household Component provides data from individual households and their members, which is supplemented by data from their medical providers. The Insurance Component is a separate survey of employers that provides data on employer-based health insurance. The Household Component (HC) collects data from a sample of families and individuals in selected communities across the United States, drawn from a nationally representative subsample of households that participated in the prior year's National Health Interview Survey (conducted by the National Center for Health Statistics). During the household interviews, MEPS collects detailed information for each person in the household on demographic characteristics, health conditions, health status, use of medical services, charges and source of payments, access to care, satisfaction with care, health insurance coverage, income, and employment.45

Table 3-1. Data sources for calculation of each cost component.

Cost component Data source Direct Medical Cost MEPS Data Files including: Full Year Consolidated Data File Medical Conditions File Prescribed Medicines File Hospital Inpatient Stays File Emergency Room Visits File Outpatient Visits File Office-based Medical Provider File Home Health Services File Other Medical Expenses File

Indirect cost: Morbidity MEPS Full Year Consolidated Data File Bureau of Labor Statistics (BLS)

Indirect cost: Mortality The National Vital Statistics System (NVSS) Bureau of Labor Statistics (BLS)

22

In this study, only the household component has been used. Each year, the selected households from the prior year's National Health Interview Survey (NHIS) form a panel. Each panel is followed for a period of two years. During these two years, each household in the panel is interviewed in five rounds. Consequently, the sample for each year of the MEPS data files includes two overlapping panels. Figure 3-1 shows this overlapping panel design.46

Sample size. When using national data, a sufficient sample size must be obtained to ensure reliable survey estimates; Medical expenditure data are highly skewed. It means that a small portion of population accounts for a large portion of expenses. Medical expenditure data from MEPS follow the same pattern. Consequently, if the sample size is not large enough, “some point estimates for particular subgroups of the population may show substantial fluctuations from one year to the next that are not statistically significant.”47(p. 2615) To address this issue, the Agency for Healthcare Research and Quality (AHRQ) has set the minimum requirement of 100 un-weighted participants per cell for producing national estimates.47

Even though MEPS annual sample size is much larger than 100, the number of participants in many of the MEPS analytic and policy relevant subpopulations of interest might be less than 100. Fortunately, several consecutive years of MEPS data files can be pooled together to improve the precision of estimates and expand the types of analyses

Panel 14

1/1/2010 1/1/2011

Round 1 Round 2 Round 3 Round 4 Round 5

Round 1 Round 2 Round 3 Round 4 Round 5

Panel 15

Figure 3-1. Overlapping design of MEPS sample.

Source: MEPS HC-138: 2010 Full Year Consolidated Data File. 2012; http://meps.ahrq.gov/mepsweb/data_stats/download_data/pufs/h138/h138doc.shtml#2581 UnitedStates. Accessed 10/15/2012.

23

possible.

For this research, data was pooled from the 2009-2010 MEPS public use files, to allow for a large enough sample size. 2009 and 2010 data were the latest available MEPS datasets at the time of study. More specifically, data files used in this study were: full year consolidated data file; medical conditions file; as well as event level files for prescribed medicines, hospital inpatient stays, emergency room visits, outpatient visits, office-based medical provider visits, home health visits, and other medical expenses from 2009 and 2010 MEPS-Household Components. The pooling was conducted according to the MEPS guidelines on pooling several years of data.48

Study sample. The study population is consisted of all survey-respondents (for the years 2009 and 2010) 18 years and older, with positive person weights. Among these, individuals who reported a diagnosis of, or had a medical event associated with anxiety disorder(s) were classified as anxiety patients. Diagnoses and events were identified using Clinical Classification (CC) code. CC code aggregates conditions and procedures into mutually exclusive and clinically homogeneous categories, using Clinical Classification Software.49 The CC code 651 is specifically assigned to anxiety disorders. The conditions included in this category are: generalized anxiety disorder, panic disorder, phobias (social phobia, specific phobia), agoraphobia, obsessive-compulsive disorder, post-traumatic stress disorder, acute stress disorder, overanxious disorder, and mixed emotional disturbances.

Data Overview

As it was mentioned earlier, three major databases were used in this study as data sources. Information collected from each of these data bases is provided in the following sections:

MEPS Data Files

The majority of data was collected from MEPS:

MEPS is a set of large-scale surveys of families, individuals, their medical providers and employers across the United States. It is jointly sponsored by the Agency for Healthcare Research and Quality (AHRQ) and the National Center for Health Statistics (NCHS), and has been conducted annually since 1996. It has three major components; the Household Component (HC), the Insurance Component (IC), and the Medical Provider Component (MPC). The MEPS-HC collects data from a nationally representative sample of the civilian non- institutionalized population of the U.S.; and is intended to provide national estimates of healthcare utilization, cost, insurance cover- age, and sources of payment.50(p. 721)

24

The following MEPS data files were used in this research:

x Full Year Consolidated Data file: Each year, the Full Year Consolidated Data file provides information collected on a nationally representative sample of the civilian non-institutionalized population of the United States for that calendar year. More specifically, this file contains the following variables: survey administration, language of interview, demographics, parent identifiers, health status, disability days, access to care, employment, quality of care, patient satisfaction, health insurance, use, income, and expenditure variables.

x Medical Conditions file: For each calendar year, the Medical Conditions file provides information on household-reported medical conditions.

x MEPS Household Component Event files: There are seven event-level files in MEPS Household Component which provide information on utilization and expenditure, due to medical conditions, by type of health services received. These files are: Prescribed Medicines file, Dental Visits file (not used in this study), Other Medical Expenses file, Hospital Inpatient Stays file, Emergency Room Visits file, Outpatient Visits file, Office-Based Medical Provider Visits file, and Home Health file.

Variables used in analyses from MEPS data files. List of variables from these data files, which were used in the analyses, are provided in Table 3-2. Some of these variables are categorical while others are continuous. List of categorical variables, along with levels of each variable, is provided in Table 3-3.

Summary variables. According to the MEPS survey design, each year interviewers collect data through three consecutive rounds (for each panel). Consequently for most measures, MEPS has information in each round (round-level variables) and also at the end of year. However, some variables are presented only at round-level and the researcher needs to summarize them into single variables representing the status of a whole year. In this section, list of summary variables used in the analyses and the way they were constructed are provided.

x Perceived health status (RTHLTH), Sick-leave pay (SICPAY), union status (UNION), and occupation category (OCCCAT): The value of summery variable would be equal to the most frequent value of round variables. If round variables all have different values, then round variable with the longest reference period represents the summery variable.

x Number of days missed work due to illness/injury (DDNWRK) and number of days stayed in bed due to illness/injury (DDBDYS): Summery variable is equal to the sum of round-level variables.

x Employment status (EMPST): In MEPS, a current main job is defined for persons who either reported they were currently employed and identified a current main

25

Table 3-2. List of variables in MEPS data files used in the current study.

Variable name Description Source DUPERSID Person ID Consolidated Data File PANEL Panel Number Consolidated Data File PERWT10F Final Person Weight, Consolidated Data File VARSTR Variance Estimation Stratum Consolidated Data File VARPSU Variance Estimation PSU Consolidated Data File SEX Sex Consolidated Data File AGELAST Person’s Age Last Time Eligible Consolidated Data File RACEX Race Consolidated Data File HISPANX Hispanic Ethnicity Consolidated Data File HIDEG Highest Degree Attained Consolidated Data File POVCAT10 Family Income as Percent of Poverty Line Consolidated Data File MARRY10X Marital Status Consolidated Data File REGION10 Census Region Consolidated Data File MSA10 MSA Status Consolidated Data File RTHLTH31 Perceived Health Status RD 31* Consolidated Data File RTHLTH42 Perceived Health Status RD 42* Consolidated Data File RTHLTH31 Perceived Health Status RD 53* Consolidated Data File TOTEXP10 Total Healthcare Expenditure Consolidated Data File ERTEXP10 Total ER Expenditure Consolidated Data File OPTEXP10 Total Outpatient Expenditure Consolidated Data File OBVEXP09/10 Total Office-Based Visits Expenditure Consolidated Data File IPTEXP10 Total Hospital Inpatient Expenditure Consolidated Data File RXEXP10 Total RX Expenditure Consolidated Data File OTHEXP10 Total Equipment/Supply Expenditure Consolidated Data File HHNEXP09/10 Total Home Health Non-Agency Consolidated Data File Expenditure HHAEXP09/10 Total Home Health Agency Expenditure Consolidated Data File DDNWRK31 # Days Missed Work RD 31 * Consolidated Data File

26

Table 3-2. (Continued).

Variable name Description Source DDNWRK42 # Days Missed Work RD 42 * Consolidated Data File DDNWRK53 # Days Missed Work RD 53 * Consolidated Data File WKINBD31 # Days Stayed in Bed RD 31 * Consolidated Data File WKINBD42 # Days Stayed in Bed RD 42 * Consolidated Data File WKINBD53 # Days Stayed in Bed RD 53* Consolidated Data File EMPST31 Employment Status RD 31* Consolidated Data File EMPST42 Employment Status RD 42* Consolidated Data File EMPST53 Employment Status RD 53* Consolidated Data File HRWG31X Hourly Wage RD 31 * Consolidated Data File HRWG42X Hourly Wage RD 42 * Consolidated Data File HRWG53X Hourly Wage RD 53 * Consolidated Data File SELFCM31 Self-Employed RD 31 * Consolidated Data File SELFCM42 Self-Employed RD 42 * Consolidated Data File SELFCM53 Self-Employed RD 53 * Consolidated Data File UNION31 Union Status RD 31 * Consolidated Data File UNION42 Union Status RD 42 * Consolidated Data File UNION53 Union Status RD 53 * Consolidated Data File SICPAY31 Paid Sick Leave RD 31* Consolidated Data File SICPAY42 Paid Sick Leave RD 42* Consolidated Data File SICPAY53 Paid Sick Leave RD 53 * Consolidated Data File NUMEMP31 Number of Employees RD 31 * Consolidated Data File NUMEMP42 Number of Employees RD 42 * Consolidated Data File NUMEMP53 Number of Employees RD 53 * Consolidated Data File OCCCAT31 Occupation Group RD 31 * Consolidated Data File OCCCAT42 Occupation Group RD 42 * Consolidated Data File OCCCAT53 Occupation Group RD 53 * Consolidated Data File INS10X Insurance Indicator Variable Consolidated Data File

27

Table 3-2. (Continued).

Variable name Description Source INSCOV10 Health Insurance Coverage Indicator Consolidated Data File MCAID10X Covered by Medicaid Consolidated Data File MCARE10X Covered by Medicare Consolidated Data File TRICR10X Covered by Tricare Consolidated Data File OTPUBA10, Covered by Other Public Insurances Consolidated Data File OTPUBB10, STAPR10 PUB10X Covered by Public Insurance Consolidated Data File CONDIDX Condition ID Medical Conditions File ICD9CODX ICD-9-CM Code For Condition Medical Conditions File CCCODEX CC Code in Conditions File Medical Conditions File RXCCC1X-3X CC Code in RX File RX File ERCCC1X-3X CC Code in ER Visit File ER visits File OPCCC1X-4x CC Code in Outpatient Visit File Outpatient Visits File IPCCC1X-4X CC Code in Hospital Inpatient File Hospital Inpatient File OBCCC1X-4X CC Code in Office-Based Visit File Office-Based Visits File

Notes: Name of variables in this table are as shown in MEPS 2010 data files. In the previous years of MEPS data files, most variables have the same names. Variables ending in 10 represent the values as of 12/31/2010. So for each year of data collection, these variables would end in XX, where XX are the last two digits of the year data was collected for; Variables ending in 31, 42, or 53 are round specific variable and the last two digits at the end specify the round in which data was collected (i.e. round 1, 2, 3 of the panel started in the current year (the first digit) or round 3, 4, 5 of the panel started in the previous year (the second digit)). * These variables haven’t been used directly in the analyses. Instead, they have been used to construct summary variables, and the summary variables were included in the analyses. PSU=Primary Sampling Unit; MSA = Metropolitan Statistical Area; RD = Round; ER = Emergency Room; RX = Prescribed Medicine; # = Number; CC = Clinical Classification.

28

Table 3-3. List of categorical variables.

Variable(s) Level Value Sex 1 Male 2 Female

AGE CATEGORYa 1 18-24 2 25-44 3 45-64 4 65+

RACE/ETHNICITYb 1 White-non Hispanic 2 Black-non Hispanic 3 Hispanic 4 Other

REGION10 1 Northeast 2 Midwest 3 South 4 West

MSA10 1 Non-MSA 2 MSA

POVCAT10 1 Poor (Poverty Category) 2 Near poor 3 Low income 4 Middle income 5 High income

MARRY10X 1 Married (Marital Status) 2 Widowed/Divorced/Separated 3 Never Married

INSCOVc 1 Uninsured (Insurance Coverage) 2 Dual Eligible 3 Medicare 4 Medicaid 5 Other Public 6 Private

SELFCM 1 Yes UNION SICPAY 2 No

29

Table 3-3. (Continued).

Variable(s) Level Value EMPST31/42/53 1 Currently employed (Employment Status) 2 Has a job to return to 3 Employed during the reference period 4 Not employed with no job to return to

OCCCAT 1 Management, business, and financial operations (Occupation Category) 2 Professional and related occupations 3 Service occupations 4 Sales and related occupations 5 Office and administrative support 6 Farming, fishing, and forestry 7 Construction, extraction, and management 8 Production, Transportation, and material moving

HIDEG 1 No degree (Education) 2 GED/High school diploma 3 Bachelor’s degree 4 Master/PhD 5 Other degree

RTHLTH 1 Excellent (Perceived Health Status) 2 Very good 3 Good 4 Fair 5 Poor

CCIfreq 0 Zero comorbidity (D’Hoore Adaptation of 1 One comorbidity Charlson Comorbidity 2 Two comorbidities Index)d 3 Three or more comorbidities

Note: The levels reported for some categorical variables here, do not exactly match the levels originally provided in MEPS data files. For some variables, we had to combine two or more levels to summarize data, or to get enough sample size in each level. a This variable was constructed using the continues variable “AGELAST”; b This variable was constructed using “RACEX” and “HISPANX” variables; c This variable was constructed using the following variables: INS10X, INSCOV10, MCAID10X, MCARE10X, TRICR10X, OTPUBA10, OTPUBB10, STAPR10, and PUB10X; d This variable was constructed using ICD-9-CM cods, according to the conditions and weights listed in the D’Hoore adaptation of CCI. GED = Graduate Equivalency Degree; MSA = Metropolitan Statistical Area.

30

job (at the time of interview), or those who reported and identified a job to return to. So, If an individual had a current main job for at least one round in year (at least one of the round-level variables was equal to 1 or 2), then he/she was considered as being employed (EMPST=1).

x Hours worked per week (HOUR), number of employees (NUMEMP), and hourly wage (HRWG): Summery variable is equal to the mean of round-level variables.

x Self- employment status (SELFCM): For employed individuals, If at least in one of the reported current main jobs, the person was self-employed, he/she is considered as self-employed (SELFCM=1).

BLS Data

“The Bureau of Labor Statistics (BLS) is an independent national statistical agency that collects, processes, analyzes, and disseminates essential statistical data to the American public, the U.S. Congress, other federal agencies, state and local governments, business, and labor. The BLS also serves as a statistical resource to the Department of Labor.”51 Examples of information that BLS provides are: employment cost trends, national compensation data, wages by area and occupation, earnings by demographics, earnings by industry, employee benefits …

The specific information collected from BLS is:

x Weekly & Hourly Earnings:52 The BLS reports (median) weekly earnings by industry type, occupation type, sex, race, ethnicity, age, education level, class of worker and labor force status. This information is based on the Current Population Survey (CPS).The CPS is a monthly survey of households (the sample represents the civilian non-institutional population of the U.S.) conducted by the Bureau of Census for the BLS. It provides a comprehensive body of data on the labor force, employment, unemployment, persons not in the labor force, hours of work, earnings, and other demographic and labor force characteristics.

x Labor Force Statistics:53 The data contains Labor Force Participation Rate (LFPR) by age, race, and gender. This information is also based on the CPS.

x Healthcare component of the Consumer Price Index(CPI) as well as CPI for all items: This information is available in the CPI Detailed Report Data for March 2013.54

NVSS Data

The National Center for Health Statistics (NCHS) is a component of the Centers for Disease Control and Prevention (CDC), and “its mission is to provide statistical

31

information that will guide actions and policies to improve the health of the American people. To carry out its mission, NCHS conducts a wide range of annual, periodic, and longitudinal sample surveys and administers the National Vital Statistics Systems.”55 The NVSS collects data on vital events including births, deaths, marriages, and divorces in the United States. The researcher used the following reports to collect required data. Number of deaths due to anxiety disorders were extracted from the first three reports, while the fourth one was used to collect information on life expectancy.

x Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 5-year age groups, by race and sex: United States, 1999-2010.56

x Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 10-year age groups, by Hispanic origin, race for non-Hispanic population and sex: United States, 2010.57

x Deaths, percent of total deaths and rank order for 113 selected causes of death and Enterocolitis due to Clostridium difficile, by Hispanic origin, race for non- Hispanic origin and sex, United States, 2010.58

x Deaths: Final data for 2010.59

Analysis Technique

In this section, the analysis technique for each specific aim of the study is explained.

SA1: Estimating the Societal Cost of Anxiety Disorders for the U.S. Adult Population

According to Equation 3-1, the societal cost of anxiety disorder(s) is equal to summation of its direct and indirect costs.

Total cost = Overall direct medical cost + Indirect cost (Eq. 3-1)

In the following sections, the methodology used to estimate each component of Equation 3-1 is explained.

Overall direct medical cost. Overall direct medical cost incorporates costs due to prescription medications, office-based medical provider visits, inpatient visits, outpatient visits, ER visits, and other medical expenses. In this section, we explain the methodology used to estimate the overall direct medical costs due to anxiety disorders. Estimating direct medical cost by category of health service (SA2), is explained in the next section.

32

To estimate overall direct medical expenditures attributable to anxiety disorders, we used the incremental cost approach. This method, suggested by the AHRQ for estimating cost of illness with MEPS data, has been widely used by previous researchers.60-66 The incremental cost approach provides an estimate of disease- attributable expenses by calculating expenditures incurred by a disease population in excess of those incurred by a disease free population. This difference in expenses by the two populations represents the total cost of illness, including treatment expenses, as well as expenses related to complications of the disease and its comorbidities.65 To adjust for potential differences in socioeconomic and clinical characteristics of the two populations, which are considered to have an impact on cost, a multivariate regression analysis is conducted. As such, this approach estimates expenditures solely associated with the disease of interest.64

To estimate direct medical costs associated with anxiety disorders, first an appropriate multivariate regression analysis needed to be developed to explain healthcare costs through a set of explanatory variables. The outcome variable of the model was the overall healthcare expenditure. Expenditure (cost) refers to what is actually paid for healthcare services and is defined as the sum of direct payments by different sources such as out-of-pocket payments, as well as payments by private insurance, Medicaid, and Medicare. Expenditures are more accurate than charges for cost estimation purposes. Charges vary from what is actually paid, due to uncollected liability, bad debt, charitable care, and implementing contract negotiations.46

The main explanatory variable of the model was a disease- indicator variable, indicating whether each individual in the sample suffered from anxiety disorder(s) or not. Other covariates of the model were respondents’ gender, age, race/ethnicity, marital status, education, poverty category, geographic region, metropolitan statistical area (MSA), perceived health status (PHS), health insurance coverage, and the D’Hoore adaptation of Charlson Comorbidity Index (CCI). The D’Hoore adaptation of CCI controls for 17 medical conditions which are myocardial infarct, congestive heart failure, peripheral vascular disease, cerebrovascular disease, dementia, chronic pulmonary disease, connective tissue disease, ulcer disease, mild liver disease, hemiplegia, moderate or severe renal disease, diabetes , any tumor, leukemia, lymphoma, moderate or severe liver disease, and metastatic solid tumor.67 These covariates were selected based on the literature review and the availability of data in MEPS.

After developing and running the regression model, the estimated coefficients of the model were used to calculate two predicted expenses for each individual. The first predicted expenses assumed the individual had anxiety disorder(s) (by setting the disease- indicator variable to one), and the second predicted expenses assumed the individual was anxiety-free (by setting the disease-indicator variable to zero). The average per-person increase in medical expenses attributable to anxiety disorders was calculated by taking the difference in predicted expenses for each person and computing the weighted average of the difference across the entire sample. Finally, the average per-person increase in expenses was multiplied by the weighted number of individuals with anxiety disorders in the sample, to get the total medical cost of anxiety disorders.

33

In modeling healthcare cost data, the specific distributional characteristics of the data must be taken into consideration. These characteristics are: (1) non-negative observations; (2) excessive zero (i.e. there are a large number of individuals with zero cost); and (3) highly skewed data (i.e. the majority of cost is incurred by a few patients).68 These characteristics make traditional Ordinary Least Square (OLS) regression inefficient in modeling healthcare cost data. Alternatively, OLS with natural log-transformed cost has been widely used to deal with heavily skewed data. However, this approach has some limitations as well.68 First, the outcome variable is the logarithm of cost. So in order to draw useful conclusions about the cost, the predicted values need to be retransformed back to the original scale. While retransforming the outcome variable, by using smearing factor, seems straightforward, interpretation of parameter estimates may still be challenging. Second, in the presence of heteroskedasticity, using one smearing factor to retransform the predicted values leads to biased estimates, i.e. under-estimation or over- estimation of the actual cost. In such scenarios, a Generalized Linear Model (GLM), with appropriate variance and link functions, is more efficient to model cost data. GLM directly models both the variance and mean functions on the original scale of dependent variable. As such, results can be interpreted with no need for retransformation from log scale to the original scale.68 “The mean function, E(y|x) is represented as μ(xcβ), where μ is the inverse link between the expectation of the observed raw-scale y and the linear predictor xcβ.”68(p. 529) The link function generally used with healthcare cost data is the log-link function.68 “Then μ is the exponential function. A commonly used family of variance functions includes the power functions of the form v(x) = N (μ(xcβ))λ.”68(p. 529) The specific type of variance functions depends on the value of λ. for instance, if the λ is equal to 1 it means that variance is proportional to meant. So, it would be a Poisson like model If the λ is equal to 2, it means that variance is proportional to mean squared, as in a Gamma-like model.68

In order to find out which type of regression model was most appropriate for our data, several diagnostics tests needed to be performed. First of all, we needed to find the distribution of cost data to see if data was really skewed or not. Then, appropriateness of using OLS regression with log-transformed data needed to be examined. This was achieved by conducting the Breusch-Pagan/Cook-Weisberg test for heteroskedasticity. One of the assumption of OLS is that variance of error terms (εj) should be constant (εj = V2; for all j). If this assumption is violated, i.e. when heteroskedasticity is present, OLS estimates are no longer the best linear unbiased estimator (BLUE). One of the major consequences of heteroskedasticity is biased standard errors, in which leads to biased inference, so results of hypothesis tests are possibly wrong. The Breusch-Pagan/Cook- Weisberg test for heteroskedasticity tests the linear heteroskedasticity.69 Their null and alternative hypotheses are as follows:

H0: error variances are all equal H1: error variances are a multiplicative function of one or more variables

In the case that heteroskedasticity was present; we needed to conduct a Park test to see which variance function was more appropriate to be used in the context of GLM. Park test was first introduced by Park in 1966, and its use is suggested in estimation of

34

the relationship between the mean and the variance.68,70 In this regard, “the squared residuals from a provisional model (GLM or log-transformed OLS) should be regressed on the predictions (ŷ) from the same model, both log transformed.”68(p. 531) Equation 3-2 shows the regression model for Park test:

2 ln ((yi- ŷi) ) = λ0 + λ1 ln (ŷi) + QI (Eq. 3-2)

Source: untin MB, Zaslavsky AM. Too much ado about two-part models and transformation?: Comparing methods of modeling Medicare expenditures. Journal of Health Economics. 5// 2004;23(3):525-542.

Morbidity cost. Indirect cost due to productivity loss (morbidity cost), have been measured using the Human Capital Approach (HCA). In the HCA, productivity loss due to an illness or injury is approximated by valuing the entire period of absence from work by average individual’s earning. Equation 3-3 shows the formula used to estimate the morbidity cost due to anxiety disorders:

ಿ ௡೔כ௪௚೔כσ೔సభ ௪௧೔ (ݔ (Eq. 3-3݊ܽܰ כ Morbidity Cost ൌ ൬ ಿ ൰ σ೔సభ ௪௧೔

Where: N = Total number of individuals in the sample. Wti = Person weight for the ith individual in the sample. Wgi = Daily wage for employed individuals and average daily wage for household services if the individual is not employed. ni = Number of missed work days (due to anxiety disorders) for employed individuals and number of days stayed in bed (due to anxiety disorders) for unemployed individuals. Nanx = Weighted number of individuals in the sample with anxiety disorder(s).

For individuals who were at paid employment for at least on round in year, information on wage rates are available in the MEPS (in both Consolidated Data file and Job file). For unemployed individuals, the period in which they had to stay in bed due to an illness or injury was valued by average wage for private household services. Industries in the private households are defined as those “engage in employing workers on or about the premises in activities primarily concerned with the operation of the household. These private households may employ individuals, such as cooks, maids, butlers, and outside workers, such as gardeners, caretakers, and other maintenance workers.”71 Average wage for private household services was obtained from the BLS.

Information on number of missed-work-days for employed individuals, (and number of days an unemployed person had to stay in bed) due to each particular condition, is not available in MEPS. Instead, MEPS collects this information for all medical conditions an individual might have had in the survey year. So, we needed to find out what portion of missed-work-days (variable WKINBD in MEPS consolidated data file) and bed days (variable DDBDYS) were due to anxiety disorders. In this regard, researcher used the same approach used to estimate the incremental cost. In other words,

35

two separate multivariate regression analyses were conducted to estimate the incremental number of missed work days/ bed days due to anxiety disorders. This approach has been used by Akazawa, Sindelar, and Paltiel in their research on estimating the productivity loss due to Influenza related illnesses.72

Outcome variables and the initial set of covariates for estimating the incremental number of work days/ bed days due to anxiety disorders are provided in Table 3-4. These covariates have been selected based on the relevant literature and availability of data in MEPS.

After developing and running regression models, the estimated coefficients of each model were used to calculate two outcomes for each individual. The first predicted outcome assumed the individual had anxiety disorder(s) (by setting the disease-indicator variable to one), and the second predicted outcome assumed that the individual was anxiety-free (by setting the disease-indicator variable to zero). The per-person increase in number of missed work days/ bed days attributable to anxiety disorders were calculated by taking the difference in predicted outcomes for each person. Morbidity cost for each individual in the study sample (per-person morbidity cost) was calculated by multiplying the predicted outcome (i.e. predicted number of missed work days due to anxiety disorders if the person was employed, and predicted number of bed days due to anxiety disorders if the person was not employed), by the average daily wage of that person.

Table 3-4. Outcome variables and the initial set of covariates for estimating the incremental number of missed work days/ bed days due to anxiety disorders.

Regression Outcome Sample for regression Initial set of covariates model variable analysis Model 1 WKINBD Employed individuals in anx,b gender, race/ethnicity, the study population age, marital status, education, (excluding self- poverty category, insurance employed persons)a coverage, MSA, region, CCI, number of employees, sick- pay, union status, occupation category

Model 2 WKINBD Unemployed individuals anx, gender, race/ethnicity, in the study population age, marital status, education, poverty category, insurance coverage, MSA, region, CCI. a Self-employed individuals were excluded, since information such as sick-pay benefit is not available for them. b anx represents the disease indicator variable. MSA = Metropolitan Statistical Area, CCI = Charlson Comorbidity Index.

36

Average morbidity cost was estimated by taking the weighted mean of per-person morbidity cost across the entire study sample. Finally, this average per-person cost was multiplied by the weighted number of individuals with anxiety disorders in the sample to get the total morbidity cost associated with anxiety disorders.

Mortality cost. Mortality cost is productivity loss due to premature deaths. In a prevalence-based cost of illness study, productivity losses are “calculated for all patients who die or become permanently disabled in the study year for that year and each year until the expected age of death.”3(p. 9) In the HCA, mortality cost is the present value of future earnings, from age of death to life expectancy. Lost future earnings due to a medical condition can be estimated knowing the total number of deaths due to that condition, and average annual wages of deceased individuals from the year they died to their life expectancies.

In calculating mortality cost, it is assumed that “people will be working and productive during their expected lifetime in accordance with the current pattern of work experience”73(p. 283) (for their age/sex/race cohort). So, no assumption needed to be made about the employment rate among deceased individuals. The researcher instead allowed the actual employment experience of each age/sex/race cohort inform the calculations. The estimated lifetime earnings (in present value terms) of a 25 year old White woman would not be the same as that of a 40 year old Black man, in part because of different LFPRs of these two cohorts. The LFPR in the BLS data already accounts for that source of difference, so no further explicit assumption or adjustment needed to be made to recognize the importance of this factor. LFPR is defined as the ratio of the civilian non- institutionalized population who are in labor force by the civilian non-institutionalized population who are eligible to be in labor force.

Another point to consider is that wages usually increase as individuals get more experienced. So, average annual wage at time of death should be inflated to consider this growth. According to BLS, 12-month increase in wages and salaries for the first quarter of 2013 was equal to 1.6%.74 So, we assumed an increase of 1.6% in annual wages too. Future annual earnings should then be discounted by an appropriate discount rate. The most common discount rate used in the relevant literature is 3%, which is recommended by the Panel on Cost Effectiveness in Health and Medicine.75 Current value of future earnings should be calculated for all individuals who died due to the medical condition, in the period of study, and then added up to get the mortality cost. In the case of anxiety disorders, the researcher needed to know the number of suicides due to this medical condition. It has been shown that 10% of suicides are due to anxiety disorders.5,10 So, we multiplied the total number of suicides buy 10% to get the number of deaths.

The formula used to estimate the mortality cost associated with anxiety disorders is shown in Equation 3-4.

ሺଵǤ଴ଵ଺ሻ௜ି஺ௗכௐכ ௅ ܯܶܥ ൌ σ௅௘ ೔ (Eq. 3-4) ஺ௗ ሺଵା௥ሻ೔షಲ೏

37

Where: MTC = Mortality cost for individuals who committed suicide due to anxiety disorders in the period of study. Ad= Age of death Le= Life expectancy at age of death by race and gender. Li= Labor force participation rate by gender and race at age i. W= Average annual wage by gender and race at age of death. r = Discount rate

SA2: Estimating the Incremental Direct Medical Expenditures Associated with Anxiety Disorders by Service Category

Incremental direct medical expenditures associated with anxiety disorders for different healthcare delivery settings were also estimated using the incremental cost approach. We followed the exact same procedure used for estimating the overall incremental cost. More specifically, several multivariate regression analyses were developed to separately model inpatient visits expenditure, outpatient visits expenditure, emergency room visits expenditure, prescription medications expenditure, office-based visits expenditure, and other medical expenses. So for each model, the dependent variable was the cost category of interest. Regression analyses were conducted on all individuals in the study sample.

For all models, the same set of covariates was used. The main independent variable of the models was the anxiety indicator variable (yes/no). Other covariates included respondents’ gender, age, race/ethnicity, marital status, education, poverty category, geographic region, metropolitan statistical area (MSA), perceived health status (PHS), health insurance coverage, and the D’Hoore adaptation of Charlson Comorbidity Index (CCI).

SA3: Estimating the Incremental Direct Medical Expenditures Associated with Anxiety Disorders for Different Sub-populations

Incremental direct medical expenditures associated with anxiety disorders were estimated for different sub-populations, using the incremental cost approach. These sub- populations were defined based on gender, age, race/ethnicity, marital status, education, poverty category, region, MSA, and insurance coverage. Several multivariate regression analyses were conducted to separately model overall healthcare cost for each group of individuals. The dependent variable in all models was the overall healthcare cost. To estimate the cost in each group, the regression analysis was conducted on that sub- population only and therefore, the covariate identifying that sub-population was excluded from the regression model. For instance, to estimate the incremental cost of anxiety disorders among males, the regression analysis was conducted on males, excluding gender from covariates of the model.

38

We broke down the costs among Blacks and Whites even further by gender, age, and geographic region (as you will see in the next chapter, cost of anxiety disorders was not statistically significant for Hispanics. So, there was no point in looking at the cost of anxiety disorders for Hispanics). The specific sub-populations in which incremental costs of anxiety disorders were estimated for, along with covariates of each model are provided in Table 3-5.

General Notes

For direct medical costs, all dollar amounts have been presented in 2013 dollars, using the healthcare component of the CPI (for all urban consumers). Also, for morbidity and mortality costs, all dollar amounts have been presented in 2013 dollars, using the CPI (for all urban consumers), for all items. The formulas used to present cost data from 2009/2010 dollars to 2013 dollars are provided below:

$2009 * (CPI2013 / CPI2009) = $2013 $2010 * (CPI2013 / CPI2010) = $2013

Where: $2009 = Cost in 2009 dollars. $2010 = Cost in 2010 dollars. $2013 = Cost in 2013 dollars.

For direct medical cost: 54 CPI2009 = Healthcare Component of CPI for the year 2009 = 379.516 54 CPI2010 = Healthcare Component of CPI for the year 2010 = 391.046 54 CPI2013 = Healthcare Component of CPI for the year 2013 = 424.154

For morbidity and mortality costs: 54 CPI2009 = CPI for all items for the year 2009 = 212.709 54 CPI2010 = CPI for all items for the year 2010 = 217.631 54 CPI2013 = CPI for all items for the year 2013 = 232.773

All statistical analyses were conducted using SAS software version 9.3 (SAS Institute, Cary, NC)76 and STATA software version 12.77

All analyses accounted for the complex survey design of MEPS to obtain national-level estimates. Furthermore, subpopulation analysis was conducted to generate results for individuals 18 years and older. Thereby, all results are projected to the ambulatory adult population of the U.S.

The p-value threshold was set at 0.05 to determine statistical significance of two- tailed tests.

39

Table 3-5. Sub-populations along with model specifications.

Covariates Gender Age Race/Ethnicity Marital Education Poverty Region MSA Health Ins PHS CCI

a s Sub-population Levels cat tatus

Gender Males, Females 9 9 9 9 9 9 9 9 9 9 Age in years 18-24, 24-44, 45-46, 65+ 9 9 9 9 9 9 9 9 9 9 Race/ethnicity White-non-Hispanic, Black-non Hispanic 9 9 9 9 9 9 9 9 9 9 Marital Status Married, Previously Married, Never married, 9 9 9 9 9 9 9 9 9 9 Education None, GED/High School Diploma, Bachelor, Master/PHD 9 9 9 9 9 9 9 9 9 9 Poverty Category Poor, Near poor, Low income, Middle income, High income 9 9 9 9 9 9 9 9 9 9 Region Northeast, Midwest, South, West 9 9 9 9 9 9 9 9 9 9 MSA Non-MSA, MSA 9 9 9 9 9 9 9 9 9 9 Insurance Coverage Uninsured, Dual Eligible, Medicaid, Medicare, Other public, 9 9 9 9 9 9 9 9 9 9 private only Race*Gender White-Male, White-Female, Black-Male, Black-Female 9 9 9 9 9 9 9 9 9 Rage*Age White- 18 to 44, White 45 &more, Black 18 to 44, Black 45 & 9 9 9 9 9 9 9 9 9 more Race*Region White-Northeast, White-Midwest, White-South, White-West 9 9 9 9 9 9 9 9 9 Black-Northeast, Black-Midwest; Black-South, Black-West Race*Gender*Age White – Male- 18 to 44, White – Male- 45 & more White – Female- 18 to 44, White – Female- 45 & more 9 9 9 9 9 9 9 9 Black – Male- 18 to 44, Black – Male- 45 & more Black – Female- 18 to 44, Black – Female- 45 & more a To estimate cost for each sub-population, sample includes individuals in that population only; PHS = Perceived Health Status; MSA = Metropolitan /statistical Area, CCI = Charlson comorbidity Index; GED = Graduate Equivalency Degree.

40

CHAPTER 4. RESULTS

Results of the current research are provided in this chapter. The first section provides descriptive statistics for the study population. In the second section, results of estimating direct medical costs associated with anxiety disorders are presented. Morbidity and mortality costs attributable to anxiety disorders are provided in third and fourth sections, respectively. Finally, summary of results, i.e. societal cost of anxiety disorders, is reported.

Throughout this chapter, all dollar amounts are presented in 2013 dollar values, unless otherwise is specified

Descriptive Statistics

Prevalence of Self-reported Anxiety Disorders in MEPS

From the total survey respondents in 2009-2010 (n = 66,148), 46,572 (weighted sum = 228,987,954) were adults with positive person weights and non-missing values on all of the independent variables, and were included in the final analysis. In 2009-2010 MEPS survey, 8.74% (weighted sum = 20,337,553) of adults (95% CI: 8.32% to 9.17%) reported being diagnosed with an anxiety disorder(s). The remaining 91.26% were considered as anxiety-free population.

Characteristics of the Study Population

Table 4-1 compares demographic characteristics of individuals with and without anxiety disorder(s) in the study population. Compared to adults with no anxiety disorder, those who have been diagnosed with this condition were more likely to be female (66.15% versus 50.15%; p < 0.001), White-non Hispanic (79.87% versus 66.77%; p < 0.001), in the age group of 45 to 64 (40.74% versus 34.19%; p < 0.001), and have one or more comorbidities (36.48% versus 27.98%; p < 0.001). Alternatively, they were less likely to lack insurance (15.30% versus 20.77%; p < 0.001). Also, the mean ± SE of CCI score was significantly higher at 0.82 ± 0.03, for sufferers of anxiety, compared to adults without anxiety disorders at 0.51 ± 0.01.

We also compared the two populations based on the 17 comorbidities used in the D’Hoore adaptation of the CCI (Table 4-2). In comparison with the anxiety-free population, adults with anxiety disorders were more likely to have congestive heart failure (1.49% versus 0.94%; p = 0.004), peripheral vascular disease (1.96% versus 1.28%; p = 0.020), dementia (0.95% versus 0.53%; p = 0.034), chronic pulmonary disease (16.02% versus 8.55%; p < 0.001), connective tissue disease (5.19% versus

41

Table 4-1. Comparison of demographic characteristics between adults with and without anxiety disorders.

Adults (unweighted n=47,388)a Anxiety disorder(s) No anxiety disorder

(unweighted n=3,610)a (unweighted n=43,778)a Characteristics % 95% CI % 95% CI p-value Gender < 0.001* male 33.85 32.10-35.59 49.85 49.29-50.42 female 66.15 64.41-67.90 50.15 49.58-50.72 Age in years < 0.001* 18-24 8.61 7.18-10.30 13.22 12.65-13.79 25-44 34.30 31.76-36.84 34.97 33.97-35.98 45-64 40.74 38.55-42.94 34.19 33.32-35.07 65 and Older 16.35 14.58-18.13 17.61 16.74-18.49 Race/ethnicity < 0.001* White non-Hispanic 79.87 77.82-81.92 66.77 64.88-68.65 Black non-Hispanic 6.97 5.88-8.06 11.92 10.64-13.21 Hispanic 8.99 7.63-10.36 14.41 12.78-16.04 Other non-Hispanic 4.17 3.25-5.09 6.90 5.88-7.92 Marital Status < 0.001* Married 47.02 44.36-49.68 53.80 52.70-54.91 Previously Marriedb 29.49 27.19-31.80 19.13 18.29-19.96 Never Married 23.48 21.17-25.79 27.07 26.24-27.90 Education < 0.001* No Degree 13.09 11.75-14.42 15.88 15.09-16.67 GED/High School Diploma 52.75 50.21-55.28 48.45 47.50-49.40 Bachelor 17.60 15.79-19.41 18.10 17.27-18.94 Master/PhD 7.10 5.83-8.38 9.41 8.71-10.12 Other Degree 9.46 8.06-10.87 8.16 7.68-8.63

42

Table 4-1. (Continued).

Adults (unweighted n=47,388)a Anxiety disorder(s) No anxiety disorder

(unweighted n=3,610)a (unweighted n=43,778)a Characteristics % 95% CI % 95% CI p-value Poverty Category < 0.001* Poor 16.53 14.87-18.20 12.18 11.46-12.89 Near Poor 4.07 3.24-4.89 4.37 4.06-4.67 Low Income 13.35 11.78-14.93 13.47 12.83-14.12 Middle Income 32.09 30.18-34.00 30.25 29.42-31.08 High Income 33.96 31.33-36.58 39.73 38.36-41.10 Region < 0.001* Northeast 18.34 15.74-20.95 18.48 17.06-19.91 Midwest 25.54 23.15-27.94 21.41 20.11-22.71 South 31.38 28.55-34.20 37.14 35.42-38.87 West 24.74 22.42-27.06 22.97 21.44-24.49 MSA 0.901 Non-MSA 16.00 12.62-19.38 15.85 13.35-18.35 MSA 84.00 80.62-87.38 84.15 81.65-86.66 Insurance Coverage < 0.001* Uninsured 15.30 13.53-17.06 20.77 19.72-21.81 Dual eligiblec 4.57 3.59-5.55 2.08 1.84-2.31 Medicaid 10.15 8.73-11.58 5.59 5.07-6.11 Medicare 18.32 16.44-20.20 16.94 16.09-17.79 Other Public 1.89 1.25-2.53 1.86 1.59-2.13 Private 49.76 47.33-52.20 52.77 51.47-54.06 CCI Score [mean (SE)] 0.82 (0.03) 0.51(0.01) <0.001* CCI Score (%) <0.001* Zero Comorbidity 63.52 61.39-65.66 72.02 71.25-72.79 One Comorbidity 23.93 22.08-25.78 20.18 19.67-20.78

43

Table 4-1. (Continued).

Adults (unweighted n=47,388)a Anxiety disorder(s) No anxiety disorder

(unweighted n=3,610)a (unweighted n=43,778) Characteristics % 95% CI % 95% CI p-value Two Comorbidity 8.74 7.56-9.92 5.63 5.29-5.97 Three or More 3.81 3.02-4.60 2.17 1.95-2.39

Notes: Unweighted numbers represent number of individuals in the sample, while weighted numbers represent projected number of individuals (i.e. national-level estimates), after controlling for the complex survey design of MEPS. CI = Confidence Interval; GED = Graduation Equivalency Degree; MSA = Metropolitan Statistical Area; CCI = Charlson Comorbidity Index; SE = Standard Error. a Sample estimates projected to 232,782,004 adults, among which 20,337,553 were classified as anxiety patients and the remaining 212,444,451 were considered as anxiety-free population. b Previously married refers to divorced, separated or widowed individuals. c Dual eligible refers to individuals who are entitled to Medicare Part A and/or Part B and also meet the eligibility requirements for Medicaid, therefore are enrolled in both programs. * p < 0.05, two-tailed.

44

Table 4-2. Comparison of CCI clinical conditions between anxiety and non-anxiety patients.

Adults (unweighted n=47,388)a Anxiety disorder(s) No anxiety disorder

(unweighted n=3,610)a (unweighted n=43,778)a Comorbidities % 95% CI % 95% CI p-value Myocardial infarction 3.31 2.51-4.10 2.92 2.65-3.18 0.318 Congestive heart failure 1.49 1.02-1.96 0.94 0.79-1.09 0.004* Peripheral vascular disease 1.96 1.34.2.57 1.28 1.12-1.44 0.020* Dementia 0.95 0.51-1.40 0.53 0.40-0.66 0.034* Cerebrovascular disease 0.28 0.02-0.55 0.18 0.11-0.25 0.381 Chronic Pulmonary disease 16.02 14.30-17.74 8.55 8.08-9.01 <0.001* Connective tissue disease 5.19 4.30-6.07 2.93 2.65-3.21 <0.001* Ulcer disease 0.91 0.55-1.26 0.43 0.32-0.54 0.002* Mild liver disease 0.74 0.41-1.07 0.48 0.38-0.57 0.058 Hemiplegia 3.04 2.29-3.79 2.18 1.94-2.41 0.009* Moderate or severe renal disease 0.59 0.27-0.90 0.39 0.28-0.48 0.163 Diabetes 12.24 10.93-13.54 10.78 10.28-11.28 0.032* Any tumor 7.06 5.87-8.26 6.10 5.69-6.52 0.110 Leukemia 0.11 0.00-0.25 0.15 0.09-0.21 0.621 Lymphoma 0.19 0.02-0.36 0.25 0.18-0.33 0.532 Moderate or severe liver disease 0.26 0.06-0.46 0.30 0.21-0.38 0.759 Metastasis solid tumor 0.65 0.34-0.97 0.45 0.36-0.54 0.168 a Sample estimates projected to 232,782,004 adults, among which 20,337,553 were classified as anxiety patients and the remaining 212,444,451 were considered as anxiety-free population. * p < 0.05, two-tailed. Source: D'Hoore W, Bouckaert A, Tilquin C. Practical considerations on the use of the Charlson comorbidity index with administrative data bases. J Clin Epidemiol. Dec 1996;49(12):1429-1433.

45

2.93%; p < 0.001), ulcer disease (0.91% versus 0.43%; p = 0.002), hemiplegia (3.04% versus 2.18%; p = 0.009), and diabetes (12.24% versus 10.78%; p = 0.032).

Direct Medical Costs Attributable to Anxiety Disorders

Preliminary Statistical Analyses

As it was mentioned in chapter 3, direct medical costs associated with anxiety disorders were estimated using the incremental cost approach. In this regard, several multivariate regression analysis were conducted to separately model overall healthcare expenditure, inpatient visits expenditure, outpatient visits expenditure, emergency room visits expenditure, prescription medications expenditure, office-based medical visits expenditure, and other medical expenses. However, we first needed to find the set of covariates for regression models, as well as the type of regression analyses used to model healthcare cost data. As such, the following preliminary analyses were conducted.

Checking for multicollinearity. As it was mentioned in chapter 3, an initial set of variables, based on the relevant literature and availability of data in MEPS, was selected to be included in all regression analyses. However, in the presence of strong multicollinearity between two or more covariates of a model, predictions may be biased. In such cases, either some covariates should be excluded from the model, or those with high collinearity should be combined into a new single index Variance Inflation Factor (VIF) is highly being used by researchers to identify multicollinearity. VIF simply measures how much a variable is contributing to the standard error in the regression. If there is severe multicollinearity between two or more variables in a model, the variance inflation factor will be very large for those variables. The general rule of thumb to identify multicollinearity is to use the cut point of 10 for VIF.78 That is, if the VIF for a particular variable is higher than 10, that variable should either be excluded from the model or be combined with other variables with high VIFs. Some researchers though find the cut point of 10 to be very conservative and instead go with lower cut points such as 3 or 4.78 The VIF cut point of 4 was selected in this study.

In order to find out if there is multicollinearity in our data, we run an OLS regression and asked for the VIF to be displayed in the output. The results are shown in Table 4-3. According to Table 4-3, the mean VIF for the set of covariates is equal to 2.03. Also; the majority of independent variables have VIFs less than 4. The only variables with high VIFs are age 65 to 85 (VIF=7.48), and Medicare (VIF=5.71). It means that there is possibly a high collinearity between these two variables (people 65 years and older are more likely to be enrolled in Medicare). However, we still can’t say that there is a severe collinearity between these two variables, since neither of the VIFs exceeds 10. Also, we cannot eliminate any of these two variables since they are all specific levels of other categorical variables (age and insurance). Finally, insurance coverage and age are shown to have significant impact on healthcare cost.79-83 Taking all these into account and after consulting with the biostatistician expert of the project,

46

Table 4-3. Results of regression analysis to check for multicollinearity.

Variable VIF Variable VIF No anxiety disordera Poora Anxiety disorders 1.06 Low income 1.70 Malea Middle Income 2.39 Female 1.05 High income 2.87 Age 18 to 24a Uninsureda Age 25 to 44 3.09 Dual eligible 1.72 Age 45 to 64 3.67 Medicaid 1.34 Age 65 and more 7.48 Medicare 5.71 Whitea Other public insurances 1.08 Black 1.32 Private insurance only 2.01 Hispanic 1.57 Northeasta Other 1.18 Midwest 1.88 Marrieda South 2.23 Previously married 1.27 West 2.08 Never married 1.73 Non MSAa No Degreea MSA 1.09 GED/high school diploma 1.90 Excellent PHSa Bachelor 1.87 Very good PHS 1.63 Master/PhD 1.53 Good PHS 1.72 Other degree 1.37 Fair PHS 1.56 Poora Poor PHS 1.30 Near poor 1.27 Charlson Comprbidity Index 1.34

Note: Mean VIF = 2.03. a Reference Category. VIF = Variance Inflation Factor.

47

we decided to keep all covariates in the model.

Finding the distribution of cost data. In modeling healthcare cost data, the specific distributional characteristics of the data must be taken into consideration. These characteristics are: (1) non-negative observations; (2) excessive zero (i.e. there are a large number of individuals with zero cost); and (3) highly skewed data (i.e. the majority of cost is incurred by a few patients).68 These characteristics make traditional Ordinary Least Square (OLS) regression inefficient in modeling healthcare cost data. Figure 4-1 shows the distribution of overall healthcare cost for the study population. As it was expected, healthcare cost data is highly skewed. So, OLS regression could not be used to model this type of data.

Alternatively, OLS with natural log-transformed cost data has been widely used to deal with heavily skewed data. However, this approach has some limitations as well.68 First, the outcome variable is the logarithm of cost. So in order to draw useful conclusions about the cost, the predicted values need to be retransformed back to the original scale. While retransforming the outcome variable, by using smearing factor, seems straightforward, interpretation of parameter estimates may still be challenging. Second, in the presence of heteroskedasticity, using one smearing factor to retransform the predicted values leads to biased estimates, i.e. under-estimation or over-estimation of the actual cost. Figure 4-2 depicts the distribution of the log-transformed overall cost for the study population. As it is shown in this figure, log-normal distribution is a perfect fit to our data and therefore, OLS with log-transformed cost data seems to be an appropriate regression model. However, we still needed to check for heteroskedasticity in log- transformed data. Table 4-4 shows the result of Breusch-Pagan / Cook-Weisberg test for heteroskedasticity. According to this result, error terms have non-constants standard deviation. In other forms, heteroskedasticity is present with log-transformed data. So, OLS regression with log-transformed cost data was also ruled out.

In such scenarios, a Generalized Linear Model (GLM), with appropriate variance and link functions, is more efficient to model cost data. GLM directly models both the variance and mean functions on the original scale of dependent variable. As such, results can be interpreted with no need for retransformation from log scale to the original scale.68 The link function generally used with healthcare cost data is the log-link function.68

In order to identify the appropriate variance function, a Park test was conducted. Result of this test, which is shown in Table 4-5, suggested that both Poisson and Gamma variance function might be appropriate for our data. We modeled overall healthcare cost data once using log-link GLM with Poisson distribution and then with log-link GLM with Gamma distribution These two models were compared based on (1) correlation between the predicted and observed expenditures (to see how well predictions of each model matched the observed data), and (2) mean squared error (as a summary of overall goodness of fit for each model). The comparison is shown in Table 4-6. Finally, a log- link GLM with Poisson distribution was selected as the best fitting model for modeling the overall healthcare expenditure, as well as all categories of expenditure.

48

Figure 4-1. Distribution of the overall healthcare cost for the study population.

Mu = Mean, Sigma = Standard Deviation.

49

Figure 4-2. Distribution of the log-transformed overall healthcare cost for the study population.

Mu= Mean, Sigma=Standard Deviation, totlog=log-transformed overall cost.

50

Table 4-4. Results of the Breusch-Pagan/Cook-Weisberg test for heteroskedasticity.

Variable Chi-square P-value totlog 360.07 < 0.0001*

Note: The null hypothesis in the Breusch-Pagan / Cook-Weisberg test is that all error terms have constant variance. Since the P-value is less than 0.05, we reject the null hypothesis. In other words, heteroskedasticity is present. totlog = log-transformed overall cost.

Table 4-5. Results of Park test.

Parameter estimate (λ) SE 95% CI p-value 1.64 0.01 1.61-1.66 < 0.0001*

Note: Since the parameter estimate for the λ is between 1 and 2, it means that both Poisson variance function (λ=1), and Gamma variance function (λ=2) could be appropriate candidates. SE = Standard Error, CI = Confidence interval. * p < 0.05, two-tailed.

Table 4-6. Comparison of Poisson and Gamma variance functions.

Variance Correlation between predicted Mean square error function and observed values (MSE) Poisson 0.39 1.19e+08 Gamma 0.34 2.07e+08

51

After deciding on the best fitting regression model, several multivariate regression analyses were conducted to separately model overall healthcare expenditure, inpatient visits expenditure, outpatient visits expenditure, emergency room visits expenditure, prescription medications expenditure, office-based medical visits expenditure, and other medical expenses. Results are provided in the following sections.

Overall Incremental Direct Medical Expenditure Attributable to Anxiety Disorders

Table 4-7 shows the result of regression analysis to estimate the overall incremental direct medical expenditure attributable to anxiety disorders. After controlling for gender, age, race/ethnicity, marital status, education, poverty category, geographic region, MSA, PHS, health insurance coverage, and the CCI, adults with anxiety disorders had 33% higher overall medical expenditure than those without anxiety disorders (parameter estimate: 1.33; p < 0.001). The adjusted annual overall incremental medical expenditure associated with anxiety disorders was estimated at $1657.52 per person (SE: $238.83; p < 0.001).The total annual incremental direct medical expenditures attributable to anxiety disorders, for the U.S. ambulatory adult population, is at $33.71 billion in 2013 US dollars. This figure was obtained by multiplying the per-capita incremental cost of anxiety ($1657.52) by the national estimated prevalence of anxiety disorders in MEPS (20.34 million persons).

Incremental Direct Medical Expenditure by Health Delivery Setting (SA2)

Results from individual regression models of the incremental expenditure attributable to anxiety disorders, by service category, are described in Table 4-8. The adjusted annual incremental medical expenditure associated with anxiety disorders by health service category are as follow: Inpatient care, estimated at $567.83 (SE: 176.65, p = 0.001) accounted for the largest proportion of the overall medical expenditures. Prescription medications at $531.83 (SE: 64.34; p < 0.001) accounted for the second largest proportion of the overall expenditures, followed by office-based medical provider visits at $362.41(SE: 79.58, p < 0.001). Although statistically significant, emergency room visits and other medical expenses together explained less than 8% of the overall medical expenditure. These cost categories were estimated at $37.02 (SE: 18.28; p = 0.043) and $80.85 (SE: 38.87, p = 0.038), respectively. Cost of outpatient visits estimated at $42.52 (SE: 54.69; p = 0.437), was not statistically significant.

Incremental Direct Medical Expenditure for Different Sub-populations (SA3)

In order to estimate the incremental direct medical expenditures associated with anxiety disorders for different sub-populations (based on gender, age, race/ethnicity, marital status, education, poverty category, region, MSA, and insurance coverage), separate multivariate regression analyses (GLM with Poisson distribution and log-link function) were conducted. To estimate the costs in each sub-population, the regression

52

Table 4-7. Results of regression analysis to estimate the overall incremental direct medical expenditure associated with anxiety disorders.

Parameter Parameter SE 95% CI p-value estimate No anxiety disorderb Anxiety disorders 1.33 0.05 1.24-1.43 < 0.001 Maleb Female 1.17 0.03 1.11-1.24 < 0.001 Age 18 to 24b Age 25 to 44 1.08 0.06 0.97-1.21 0.174 Age 45 to 64 1.51 0.09 1.34-1.71 < 0.001 Age 65 and Older 1.33 0.11 1.13-1.57 0.001 Whiteb Black 0.94 0.04 0.88-1.02 0.113 Hispanic 0.74 0.03 0.68-.80 < 0.001 Other 0.71 0.04 0.64-0.78 < 0.001 Marriedb Previously 0.99 0.03 0.93-1.06 0.873 marriedc Never married 0.91 0.04 0.84-0.98 0.016 No Degreeb GED/high school 1.17 0.05 1.07-1.27 0.001 diploma Bachelor 1.39 0.08 1.25-1.55 < 0.001 Master/PhD 1.42 0.09 1.26-1.61 < 0.001 Other degree 1.25 0.08 1.11-1.41 < 0.001 Poorb Near poor 0.93 0.05 0.83-1.05 0.238 Low income 1.02 0.05 0.93-1.12 0.689 Middle Income 1.02 0.05 0.93-1.12 0.687 High income 1.16 0.06 1.04-1.30 0.006 Uninsuredb Dual eligibled 3.63 0.35 3.00-4.40 < 0.001 Medicaid 2.28 0.17 1.97-2.63 < 0.001 Medicare 2.92 0.23 2.50-3.41 < 0.001 Other public 2.10 0.18 1.77-2.50 < 0.001 Private 1.93 0.12 1.71-2.17 < 0.001 Northeastb Midwest 1.04 0.05 0.96-1.14 0.345 South 0.94 0.04 0.86-1.02 0.136 West 0.99 0.04 0.91-1.08 0.759

53

Table 4-7. (Continued).

Parameter Parameter SE 95% CI p-value estimate Non MSAb MSA 1.14 0.04 1.06-1.23 0.001 Excellent PHSb Very good PHS 1.36 0.06 1.25-1.49 < 0.001 Good PHS 2.02 0.09 1.86-2.20 < 0.001 Fair PHS 3.12 0.18 2.78-3.51 < 0.001 Poor PHS 4.50 0.32 3.91-5.18 < 0.001 CCI 1.19 0.01 1.17-1.21 < 0.001 Intercept 627.05 63.76 513.14-766.24 < 0.001

Notes: Results are based on generalized linear model with log-link function and Poisson distribution. Unweighted number of individuals = 46,572. SE = Standard Error; CI = Confidence Interval; GED= Graduation Equivalency Degree; MSA = Metropolitan Statistical Area; PHS= Perceived Health Status; CCI = Charlson Comorbidity Index. a Sample estimates projected to 228,987,954 adults with positive person weights and non- missing values on all of the independent variables; b Reference Category; c Previously married refers to divorced, widowed, or separated individuals; d Dual eligible refers to individuals who are entitled to Medicare Part A and/or Part B and also meet the eligibility requirements for Medicaid, therefore are enrolled in both programs.

Table 4-8. Results of regression analyses to estimate the incremental expenditures of anxiety disorders by service category.

Service category AIC ($) SE p-value Total costa %b Inpatient visits 567.83 176.65 0.001 $11.55 B 35.00% Outpatient visits 42.52 54.69 0.437 $864.75 M 2.62% Office-based visits 362.41 79.58 <0.001 $7.37 B 22.34% Emergency room visits 37.02 18.28 0.043 $752.90 M 2.28% Prescription medications 531.83 64.34 <0.001 $10.82 B 32.78% Other medical expenses 80.85 38.87 0.038 $1.64 M 4.98% Overall expenditure 1657.52 238.83 <0.001 $33.71 B 100.00%

Note: All costs are presented in 2013 US dollars. AIC = Average Incremental Cost; SE = Standard Error; B = Billion; M = Million. a Calculated by multiplying the average incremental cost by the prevalence of anxiety disorders in MEPS (i.e. 20.34 million persons). b Percent of overall expenditure.

54

analysis was conducted on that sub-population only and therefore, the covariate identifying that sub-population was excluded from the regression model. For instance, to estimate the incremental cost of anxiety disorders among males, the regression analysis was conducted on males, excluding gender from covariates of the model. Results are provided in Table 4-9.

According to these findings, having anxiety disorder(s) leads to statistically significant increase in direct medical expenditures almost for all sub-populations. The only exceptions are Hispanics, individuals with education levels lower than GED, and those who are uninsured, have Medicaid, or other public insurances (such as VA, Tricare,…). The highest increases in direct medical costs as a result of anxiety disorder(s) were seen in the following sub-populations: dual-eligible, those who live in non-MSA regions, highly educated individuals (with Master or PhD degree), seniors (65 years of age and older), Medicare enrollees, Midwest residents, divorced, widowed, or separated individuals, females, people with middle income, and Whites (non-Hispanic).

We have looked at the cost of anxiety disorders among Blacks and Whites (but not Hispanics, since presence of anxiety disorders did not have a statistically significant impact on healthcare cost of this population) even further. More specifically, we broke down each of these race/ethnicity categories, by age, gender and geographic region. The totals of 24 separate multivariate regression analyses were conducted to estimate cost of anxiety disorders in each of these groups. Results are provided in Table 4-10. According to these results, for both race/ethnicity groups (White non-Hispanics and Black non- Hispanics) females and individuals 45 years and older accounted for the majority of cost. This is the exact same observation we had when looking at the costs of all race/ethnicities together.

Regarding geographic region, we found that geographic variations did not impact cost of anxiety disorders among White non-Hispanics. In fact, in all geographic regions, anxiety disorders led to statistically significance increase in direct medical expenditures among White non-Hispanics. More specifically, White non-Hispanics who reside in Midwest had higher costs (similar to what we had already found for all race/ethnicities combined). However, Blacks had different cost pattern. In fact, among Black non- Hispanics, only those who reside in northeast had higher medical costs due to anxiety disorders and for other geographic regions, anxiety related costs were not statistically significant. It should be further investigated to see whether prevalence of anxiety disorders is higher among Blacks in northeast, or the zero cost of anxiety disorders for people of color, who reside in other regions, is due to some barrier in receiving the required care.

These findings all demonstrate why it is important to calculate values for subgroups. If we just looked at the incremental costs for men among Whites and Blacks, it looks like the costs are very similar ($1213.60 and. $1216.57) (Table 4-10). However, looking at the age segments, we saw that the costs for White non-Hispanic males were similar between age groups ($1172.83 and $1116.29), but they were incredibly different for Black non-Hispanic males, where older Black males had an incremental cost nearly

55

Table 4-9. Incremental direct medical expenditures associated with anxiety disorders for different sub-populations.

Adjusted mean Sub-population SE p-value incremental cost ($) Gender Male 963.97 342.64 0.005* Female 2137.98 330.96 < 0.001* Age in years 18-24 1498.39 439.97 0.001* 25-44 771.47 281.03 0.006* 45-64 2102.74 432.29 < 0.001* 65 and more 2426.40 765.072 0.002* Race/ethnicity White non-Hispanic 1879.31 310.15 < 0.001* Black non-Hispanic 1459.30 671.15 0.030* Hispanic 472.98 344.68 0.170 Marital Status Married 1522.56 370.62 < 0.001* Previously Marrieda 2305.23 588.36 < 0.001* Never Married 1814.21 393.55 < 0.001* Education None 1069.70 560.54 0.056 GED/High School 1795.34 324.47 < 0.001* Diploma Bachelor 1615.17 552.84 0.003* Master/PHD 2463.31 1058.67 0.020* Poverty Category Poor 1710.80 555.70 0.002* Near Poor 1594.02 1089.02 0.143 Low Income 1087.15 601.07 0.070 Middle Income 2048.08 423.33 < 0.001* High Income 1469.45 473.18 0.002* Region Northeast 1463.21 495.22 0.003* Midwest 2338.59 572.02 < 0.001* South 1197.91 364.88 0.001* West 1718.05 421.48 < 0.001* MSA Non-MSA 2485.64 621.19 < 0.001* Insurance Coverage Uninsured 483.63 313.77 0.123

56

Table 4-9. (Continued).

Adjusted mean Sub-population SE p-value incremental cost ($) Dual Eligibleb 6455.84 1990.81 0.001* Medicaid 1129.83 769.44 0.142 Medicare 2354.90 778.10 0.002* Other Public 781.37 891.45 0.381 Private Only 1571.62 319.97 < 0.001*

Note: All costs are presented in 2013 US dollars. a Previously married refers to divorced, widowed, or separated individuals. b Dual eligible refers to individuals who are entitled to Medicare Part A and/or Part B and also meet the eligibility requirements for Medicaid, therefore are enrolled in both programs. SE = Standard Error; GED= Graduation Equivalency Degree; MSA = Metropolitan Statistical Area. * p < 0.05, two-tailed

57

Table 4-10. Breaking down the cost of anxiety disorders among different race/ethnicities by gender, age, and geographic region.

Incremental direct medial cost (SE) Sub-population White non-Hispanic Black non-Hispanic Gender Male 1213.60 (475.32)* 1216.57 (920.13) Female 2318.78 (424.48)* 1797.20 (865.29)* Agea 18-44 1060.49 (307.34)* 1268.23 (803.82) 45 and over 2431.47 (497.92) * 2189.18 (995.05) * Region Northeast 1406.38. (532.84) * 3899.40 (1890.84) * Midwest 2071.76 (600.52) * 824.05 (1054.74) South 1413.00 (435.31) * 491.19 (645.86) West 1478.93 (450.03) * 2001.25 (1459.93) Gender - Age Male - 18 to 44 1172.83 (444.11) * 650.61 (417.22) Male - 45 and over 1116.29 (738.16) 3932.22 (1767.89) * Female - 18 to 44 1235.21 (456.88) * 2315.74 (1277.64) Female - 45 and over 3150.09 (696.68) * 1843.72 (1314.72)

Note: All costs are presented in 2013 U.S. dollars; Results are not provided for the Hispanics, since presence of anxiety disorder(s) did not have a statistically significant impact on healthcare cost of this population. a Age groups were combined to get enough sample size in each cell. SE = Standard Error. * p < 0.05, two-tailed.

58

six times that of the 18-44 year old ($3932.22 and $650.61). For women, we saw a difference in the costs by age group, but older White women had a higher cost (nearly as much as older Black men) and for Black women we saw higher cost is in the younger age group. So, there is a need for future studies to figure out why all of these incremental costs vary so much.

Morbidity Cost Associated with Anxiety Disorders

As it was mentioned in the third chapter, morbidity cost was measured using the Human Capital Approach (HCA). In the HCA, productivity loss due to an illness or injury is approximated by valuing the entire period of absence from work by average individual’s earning. For individuals who are at paid employment for at least one round in the year, information on wage rates are available in the MEPS (in both Consolidated Data file and Job file). For unemployed individuals, the period in which they had to stay in bed due to an illness or injury is valued by the average wage for private household services. Equation 4-1 shows the formula to calculate the morbidity cost.

ಿ ௡೔כ௪௚೔כσ೔సభ ௪௧೔ (ݔ (Eq. 4-1݊ܽܰ כ Morbidity Cost ൌ ൬ ಿ ൰ σ೔సభ ௪௧೔

Where: N = Total number of individuals in the sample. Wti = Person weight for the ith individual in the sample. Wgi = Daily wage for employed individuals and average daily wage for household services if the individual is not employed. ni = Number of missed work days (due to anxiety disorders) for employed individuals and number of days stayed in bed (due to anxiety disorders) for unemployed individuals. Nanx = Weighted number of individuals in the sample with anxiety disorder(s).

Number of Missed Work Days due to Anxiety Disorders for Employed Individuals

Since MEPS reports the total number of missed work days for each individual (due to all medical conditions he/she might have), we needed to find the incremental number of missed work days specifically due to anxiety disorders. A multivariate regression analysis was conducted (the very same approach we used to estimate the incremental direct medical costs). The outcome variable was the total number of missed work days and the covariates were gender, age, race/ethnicity, marital status, education, geographic region, MSA, Charlson comorbidity index, occupation category, union status, number of employees, and sick pay (whether or not the person has the sick pay benefit).

Checking for multicollinearity. Table 4-11 shows the results of multicollinearity analysis. VIF for insurance coverage and poverty category were estimated at 5.78 and 4.91 respectively. Since both of these values exceeded the cut point of 4, and also there is no evidence in the literature supporting the impact of these

59

Table 4-11. Results of multicollinearity analysis for modeling the number of missed work days.

Variable VIF Variable VIF No anxiety disordera Management, business, and financial operationsa Anxiety disorders 1.03 Professional and related occupations 2.31 Malea Service Occupations 2.37 Female 1.22 Sales and related occupations 1.75 Age 18 to 24a Office and administrative support 2.11 Age 25 to 44 3.53 Farming, fishing, forestry 1.14 Age 45 to 64 3.97 Construction. Extraction, and maintenance 1.64 Age 65 to 85 1.44 Production, transportation 2.06 Whitea Sick-pay benefita Black 1.24 No sick-pay benefit 1.34 Hispanic 1.30 Uniona Other 1.12 Non-union 1.13 Marrieda Number of employees 1.11 Previously married 1.19 Northeasta Never married 1.46 Midwest 2.10 No Degreea South 2.35 GED/high school diploma 3.08 West 2.14 Bachelor 3.03 Non MSAa Master/PhD 2.35 MSA 1.06 Other degree 1.95 Charlson Comprbidity Index 1.08

Note: Mean VIF = 1.85. a Reference Category. VIF = Variance Inflation Factor.

60

variables on the number of missed work days,72 they were excluded from the model. There is no multicollinearity with the remaining set of covariates.

Finding the distribution of missed work days. Figure 4-3 shows the distribution of number of missed work days (“wkinbd” variable in the MEPS data file). Since the “wkinbd” had a highly-skewed distribution, use of OLS regression would have led to biased estimates. So, OLS with log-transformed data and GLM regression were considered. OLS with log-transformed data was ruled out; due to the heteroskedasticity in data (result of the test for heteroskedasticity is provided in Table 4-12). Therefore, we conducted the park test to identify the best variance function to be used in the GLM regression (Table 4-13).

According to the results of Park test, both Poisson and Gamma variance functions were appropriate fit to our data. We conducted a. log-link GLM with Poisson distribution (to be consistent with the methodology used in estimating the direct medical costs), on the employed individuals 18 years and older. Self-employed individuals were not included in the analysis since information such as sick leave is not provided for them in the MEPS. Table 4-14 shows the result of regression analysis to estimate the incremental number of days missed work due to anxiety disorders. Based on these results, having anxiety disorder(s) has increased the number of missed work days by almost 2.5 days.

Number of Days Stayed in Bed due to Anxiety Disorders for Unemployed Individuals

For unemployed individuals, MEPS collects the total number of days each individual had to stay in bed (due to all medical conditions he/she might have had during the survey year). The variable representing this information in the MEPS consolidated data file is called “ddbdys”.we needed to find the incremental number of bed days specifically due to anxiety disorders. Distributional characteristics of the variable “ddbdys” were pretty the same as they were for the variable “wkinbd”. In other words, “ddbdys” was also highly skewed and error terms from the OLS regression on log- transformed “ddbdys” were also heteroskedastic. So, we conducted a log-link GLM with Poisson distribution on the unemployed individuals in the sample to estimate the incremental number of bed days due to anxiety disorder (s). The dependent variable in the regression model was “ddbdys” and independent variables were gender, age, race/ethnicity, education, marital status, geographic region, MSA, and Charlson comorbidity index. Poverty category and insurance coverage were excluded from the set of covariates due to their high multicollinearity with other variables (high VIF). Table 4-15 shows the result of regression analysis to estimate number of bed days due to anxiety disorders for unemployed individuals. As one can see, having anxiety disorders has increased the number of bed days by more than 12 days, for unemployed individuals.

61

Figure 4-3. Distribution of the number of missed work days for employed individuals.

WKINBD = Number of missed work days.

62

Table 4-12. Results of the Breusch-Pagan/Cook-Weisberg test for heteroskedasticity.

Variable Chi-square p-value wkinbd_log 218.33 < 0.0001*

Note: The null hypothesis in the Breusch-Pagan / Cook-Weisberg test is that all error terms have constant variance. Since the P-value is less than 0.05, we reject the null hypothesis. In other words, heteroskedasticity is present. wkinbd_log = log-transformed missed work days.

Table 4-13. Results of Park test.

Parameter estimate (λ) SE 95% CI p-value 1.90 0.04 182-1.98 < 0.0001*

Note: Since the parameter estimate for the λ is between 1 and 2, it means that both Poisson variance function (λ=1), and Gamma variance function (λ=2) could be appropriate candidates. SE = Standard Error, CI = Confidence interval. * p < 0.05, two-tailed.

Table 4-14. Incremental number of missed work days due to anxiety disorders.

Incremental estimate SE 95% CI p-value 2.18 0.57 1.05,3.30 < 0.0001*

SE = Standard Error, CI = Confidence interval. * p < 0.05, two-tailed.

Table 4-15. Incremental number of bed days due to anxiety disorders.

Incremental estimate SE 95% CI p-value 12.55 1.62 9.38,15.72 < 0.0001*

SE = Standard Error, CI = Confidence Interval. * p < 0.05, two-tailed.

63

Daily Wage for Employed Individuals

For individuals who were employed for at least one round during the survey year, MEPS collects information on hourly wages, as well as the number of hours a person works during a week. To convert hourly wages to daily wages, we assumed that each individual worked 5 days a week. So, daily wage for employed individuals was calculated according to the Equation 4-2.

DW= hours worked per day* HW = (hours worked per week/5) * HW (Eq. 4-2)

Where: DW = Daily wage. HW = Hourly wage as it comes in the MEPS consolidated data file.

Average Daily Wage of Household Services for Unemployed Individuals

As it was mentioned in the chapter 3, in the HCA, morbidity cost for unemployed individuals is calculated by valuing the time period in which an individual had to stay in bed, due to an illness or injury (bed days).the assumption is that daily wage for unemployed individuals is equal to the daily wage for services usually being done at home (household services). For each survey year (2009 and 2010), we used the average daily wage for cooks, maids and maintenance services, as the average daily wage for household services. Results are provided in Table 4-16.

Summary of Analyses

Having all the information required to calculate the morbidity cost attributed to anxiety disorders (according to Equation 4-1), summary of results are provided in Table 4-17. The adjusted mean incremental morbidity cost due to anxiety disorders was estimated at $625.73 in 2013 US dollars. Multiplying this figure by the weighted number of individuals with anxiety disorder(s) in this period (i.e. 20,337,553 adults), the total morbidity cost associated with this category of mental illnesses was estimated at $12.72 billion in 2013 US dollars.

Mortality Costs Associated with Anxiety Disorders

As it was already defined in the chapter 3, mortality cost is productivity loss due to premature deaths. In the HCA, mortality cost is the present value of future earnings, from age of death to life expectancy. In this regard, mortality cost in HCA is calculated using the formula in Equation 4-3.

ሺଵǤ଴ଵ଺ሻ௜ି஺ௗכௐכ ௅ ܯܶܥ ൌ σ௅௘ ೔ (Eq. 4-3) ஺ௗ ሺଵା௥ሻ೔షಲ೏

64

Table 4-16. Average daily wage for unemployed individuals based on BLS data.

Weekly wage ($)a Daily wage ($)b Gender Service category 2009 2010 2009 2010 Men Cook 400 401 Maid 444 455 Maintenance 488 493 Average 444 450 97 96 Women Cook 371 381 Maid 371 376 Maintenance 388 391 Average 377 383 82 82

Note: Wage data for specific service categories is not available by further details such as race/ethnicity. a In the current dollar value; b In 2013 dollar value.

Table 4-17. Summary of results: Morbidity cost.

Adjusted incremental Cost SE 95% CI p-value estimate ($) Average 625.73 7.40 611.14,640.32 < 0.0001* Total 625.37*20,337,553= $12.72 billion in 2013 US dollars

SE = Standard Error, CI = Confidence interval. * p < 0.05, two-tailed.

65

Where: MTC = Mortality cost for individuals who committed suicide due to anxiety disorders in the period of study. Ad= Age of death. Le= Life expectancy at age of death by race/ethnicity and gender. Li= Labor force participation rate by gender and race/ethnicity at age i. W= Average annual wage by gender and race/ethnicity at age of death. Discount rate is equal to 3%.

Number of People Who Committed Suicide due to Anxiety Disorders and Age at Death

NVSS provides number of deaths by age group, gender, Hispanic origin, and race for non-Hispanic origin for different causes of death. These statistics are reported in several different formats, based on the age group (5-year age group or 10-year age group) and causes of death (for 15 major causes of death or for 113 causes of death).56-58 We started collecting the number of deaths due to suicides for 2010, using the number of deaths provided by 15 major causes of death in 5-year age groups, by Hispanic origin, race for non-Hispanic population and sex. However, if for some specific sub-populations suicide did not appeared in the first 15 causes of death, we had to use other data tables to collect as much information as possible. Number of deaths due to suicide by age, gender, and race/ethnicity are provided in Table 4-18. You may see that for some sub- populations the number of suicides is provided for 5-year age groups and for others in 10- year age group. Since we had the number of deaths for each age group but not at each specific age, we assumed that all deaths in each age group (either 5 or 10 year age groups) have occurred in the middle point of that range. For instance, we assumed that for all deaths between the ages of 20 to 25, the age at death was 22. In the same way, we assumed that for all deaths occurred between the ages 65 to 75, the age at death was 70.

It has been shown that 10% of suicides are due to anxiety disorders.5,10 So, to get the number of suicide due to anxiety disorders at certain age by gender and race/ethnicity, we multiplied the figures in Table 4-18 by 10%.

Life Expectancy at Age of Death by Race/Ethnicity and Gender

NVSS also provides statistics on life expectancy at age of death by race/ethnicity and gender.59 This information is provided in Table 4-19. If the age at death was a multiplication of 10 (for 10-year age group), then we already have the life expectancy at that age. However, for 5-year age groups (for instance 20 to 25), the life expectancy at the age of death (22) is equal to the mean of life expectancies at the beginning (20) and end (25) of that range.

66

Table 4-18. Number of suicides in 2010 by age, gender, and race/ethnicity.

White Black Hispanic Age Male Female Male Female Male Female group n % n % n % n % n % n % 20-24 1,717 84% 335 16% 259 84% 49 16% 307 82% 68 18% 25-29 1,822 82% 410 18% 239 85% 43 15% 272 84% 51 16% 30-34 1,658 79% 445 21% 186 88% 26 12% 232 88% 33 12% 35-39 1,887 77% 567 23% 180 79% 47 21% 211 78% 59 22% 40-44 2,213 76% 692 24% 160 77% 48 23% 196 83% 39 17% 45-49 2,928 77% 878 23% 140 80% 36 20% 197 82% 42 18% 50-54 2,996 76% 953 24% 119 76% 37 24% 150 82% 33 18% 55-59 2,549 76% 821 24% 97 48% 138 83% 29 17% 36 18% 60-64 1,806 76% 558 24% 70 34% 67 78% 19 22% 65-69 1,210 79% 322 21% 57 78% 16 22% 66 94% 4 6% 70-74 972 84% 179 16% 40 78% 11 22% 75-79 878 47% 246 13% 38 79% 10 21% 59 89% 7 11% 80-84 761 40% 85+ 791 88% 110 12% 9 75% 3 25% 29 97% 1 3% Total 24,188 79% 6,516 21% 1,563 82% 339 18% 1,955 83% 408 17%

Sources: Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 10-year age groups, by Hispanic origin, race for non-Hispanic population and sex: United States, 2010. Mortality Tables 2012; http://www.cdc.gov/nchs/nvss/mortality_tables.htm. Accessed 11/25/2012 ; Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 5-year age groups, by Hispanic origin, race for non-Hispanic population and sex: United States, 2010. Mortality Tables 2012; http://www.cdc.gov/nchs/nvss/mortality_tables.htm. Accessed 11/25/2012; Deaths, percent of total deaths and rank order for 113 selected causes of death and Enterocolitis due to Clostridium difficile, by Hispanic origin, race for non-Hispanic origin and sex, United States, 2010, Mortality Tables 2012.

67

Table 4-19. Life expectancy at selected ages by race, Hispanic origin, race for non- Hispanic population, and sex: United States, 2010.

White non- Black non- Hispanic Hispanic Hispanic Age at Male Female Male Female Male Female death 20 59.3 64.4 57.2 61.1 52.9 58.8 25 54.6 59.5 52.5 56.9 48.4 54.0 30 49.8 54.6 47.9 52.0 43.9 49.2 35 45.1 49.7 43.2 47.2 39.4 44.5 40 40.4 44.8 38.6 42.5 34.9 39.8 45 35.7 40.0 34.0 37.8 30.5 35.3 50 31.2 35.3 29.7 33.2 26.3 31.0 55 26.6 30.8 25.5 28.8 22.5 26.8 60 22.8 26.3 21.5 24.4 19.0 22.9 65 18.8 22.0 17.7 20.3 15.8 19.1 70 15.1 18.0 14.2 16.4 12.8 15.7 75 11.7 14.1 11.0 12.8 10.1 12.5 80 8.7 10.7 8.1 9.6 7.8 9.6 85 6.1 7.7 5.8 6.9 5.9 7.1 90 4.2 5.4 4.0 4.8 4.4 5.2 95 2.9 3.7 2.8 3.3 3.3 3.8 100 2.1 2.6 2.1 2.3 2.6 2.8

Source: Murphy SL, Xu JQ, Kochanek KD. Deaths: Final Data for 2010. National Vital Statistics Reports. Hyattsville, MD: National Center for Health Statistics;2013.

68

Labor Force Participation Rate and Wage

We collected information on LFPR and wage from the BLS.52,53 This information is provided in Tables 4-20 and 4-21 respectively. In BLS, wage data are available only for age groups of 16-24, 25-54, and 55+. So, we could not break down the wage data into finer age groups.

Summary of Analyses

Having all the information required to calculate mortality cost according to Equation 4-3, results of this section are provided in Table 4-22. Assuming that 10% of suicides are due to anxiety disorders, 3,497 individuals committed suicide in 2010, with anxiety disorder(s) as primary reason. This resulted in $2.34 billion (in 2013 US dollars) loss in terms of mortality cost. White-non Hispanics accounted for almost 90% of this figure, mainly because of significantly higher number of suicides in this sub-population. Among White-non Hispanics, males between the ages of 25 and 44 years, had the highest mortality cost (37.71% of total mortality cost).

The Societal Cost of Anxiety Disorders for the U.S. Adult Population in 2010 (SA1)

The societal cost of anxiety disorders was estimated, by adding-up the overall direct medical cost, morbidity cost, and mortality cost, at almost $49 billion in 2013 US dollars. Direct medical cost accounted for the majority of this figure (69.12%), followed by morbidity cost at 26.08% and mortality cost at 4.81%. These results are provided in Table 4-23.

69

Table 4-20. Labor force participation rate by age, gender, and race/ethnicity.

White non-Hispanic Black non-Hispanic Hispanic Age Men Women Men Women Men Women group 20-24 77.0 69.7 66.9 66.9 80.0 61.6 25-29 90.0 76.2 82.3 76.7 91.9 67.7 30-34 92.3 73.6 84.7 79.3 93.5 64.9 35-39 93.3 73.7 86.8 78.2 94.1 66.7 40-44 91.6 76.2 85.5 77.1 91.5 69.3 45-49 89.7 77.2 79.6 75.3 88.6 72.2 50-54 86.5 75.3 75.1 70.6 86.7 67.7 55-59 79.7 69.4 65.2 63.6 77.1 60.5 60-64 61.3 51.7 46.7 44.2 57.8 44.5 65-69 37.2 27.6 27.9 24.2 38.7 24.3 70-74 22.5 15.0 16.3 13.0 23.4 10.4 75+ 10.5 5.3 9.3 5.6 10.9 5.5

Source: Labor Force Statistics from the Current Population Survey. Bureau of Labor Statistics. http://bls.gov/data/. Accessed 11/05/2012.

Table 4-21. Median usual weekly earnings in current dollar from current population survey for 2010.

White non-Hispanic Black non-Hispanic Hispanic Age group Men Women Men Women Men Women 16-24 453 424 403 404 395 392 25-54 878 715 656 615 587 529 55+ 990 734 740 614 617 521

Notes: Wage data are in 2010 US dollars; in converting weekly wages to annual wages, the assumption was that each year is consisted of 52 weeks. Source: Weekly and Hourly Earnings from the Current Population Survey Bureau of Labor Statistics. http://bls.gov/data/. Accessed 11/05/2012

70

Table 4-22. Mortality cost: Summary of results.

White non-Hispanic Black non-Hispanic Hispanic Age %/$a Male Female Both sexes Male Female Both sexes Male Female Both sexes group $ 247.09 M 32.23 M 279.33 M 24.46 M 3.99 M 28.45 M 29.62 M 4.36 M 33.98 M 18-24 % 10.54 1.38 11.92 1.04 0.17 1.21 1.26 0.19 1.45 $ 883.91 M 159.56 M 1.04 B 58.92 M 10.31 M 69.23 M 71.43 M 9.31 M 80.75 M 25-44 % 37.71 6.81 44.52 2.51 0.44 2.95 3.05 0.40 3.45 $ 591.12 M 114.36 M 705.48 M 14.25 M 3,06 M 17.31 M 21.1 M 2.90 M 23.99 M 45-65 % 25.22 4.88 30.10 0.61 0.13 0.74 0.90 0.12 1.02 $ 33.50 M 26.63 M 60.13 M 500,945 27,077 528,022 988,306 112,941 1.10 M 65+ % 1.43 1.14 2.57 0.02 0.00 0.02 0.04 0.00 0.05 All $ 1.76 B 332.79 M 2.09 B 98.13 M 17.34 M 115.52 M 123.14 M 16.67 M 139.82 M Ages % 74.91 14.20 89.11 4.19 0.74 4.93 5.25 0.71 5.97 Total mortality cost in 2013 US dollar 2,343,749,010 a $ represents the actual cost in 2013 US dollars, while % represents the share of cost in each cell from the total mortality cost, in terms of percentage. For instance, White- non Hispanic males between the ages of 18 and 24, had $247.09 million mortality cost, which accounts for 10.54% of the total mortality cost. M = Million; B = Billion.

Table 4-23. The societal cost of anxiety disorders for the U.S. adult population in 2010.

Indirect cost Overall direct medical cost Morbidity cost Mortality cost Societal cost $33,709,900,849 (69.12%) $12,718,495,520 (26.08%) $2,343,749,010 (4.81%) $48,772,145,379

Note: Costs are in 2013 US dollar values.

71

CHAPTER 5. DISCUSSION

Direct Medical Costs

General Notes

Our results showed that almost 9% of the ambulatory adult population of the U.S. in 2009-2010 was diagnosed with anxiety disorder(s). This estimate is lower than the 18.1% estimate for the 12-month prevalence of any anxiety disorder, reported by the National Comorbidity Survey Replication (NCS-R).9 This difference could be explained by the fact that in MEPS, medical conditions are self-reported; while in the NCS-R, a fully structured diagnostic interview was conducted to identify individuals with a particular mental disorder. So, it is possible that some individuals with mild or moderate anxiety disorder did not receive the diagnosis at the time of interview. Another possibility is that some persons did not report their anxiety, simply because of the social stigma attached to mental disorders. All these can lead to under-representation of anxiety disorders in MEPS. Consistent with the NCS-R’s findings, this study also found that prevalence of anxiety disorders is higher among women.

Unlike the previous researchers, who collected data from several different data sources, only one database was used in the current study to estimate direct medical costs. Using multiple data sources may lead to data inconsistency. To avoid this issue, we used MEPS as the only data source for all analyses related to direct medical costs, since it contains comprehensive information regarding the health care utilization and cost for participants in the survey.

MEPS has been widely used by previous researchers for the estimation of disease- attributable expense.60-64,66,84-86 In fact, when it comes to highly prevalent diseases, MEPS is superior to administrative databases for cost estimation purposes in several ways. First of all, it contains detailed information on demographic and clinical characteristics of individuals, as well as their health care utilization and expenses. So, by controlling for all factors that may affect healthcare expenditures in analysis, researchers can estimate costs solely due to the condition of interest. Second of all, disease-attributable expenses can be presented as point estimates, as well as percentage of total costs of the disease population. This information, which cannot be gained using administrative data, provides a more sensible picture of the economic burden of a disease;65 Finally, MEPS is the only database which contains all required information to estimate direct medical expenditures attributable to a disease. So instead of getting data from several different data sources, which may lead to data inconsistency and later cast doubt on the reliability of results, only one database is used to collect data on healthcare utilization and costs.

The confidence in the current findings is also derived from the adaptation of a robust statistical analysis technique. In modeling healthcare cost data, an attempt was

72

made to reduce bias in estimates by selecting statistical techniques that considered the skewed nature of such data.

Overall Direct Medical Cost

Using estimates from this study, the annual incremental direct medical expenditures attributable to anxiety disorders, for the U.S. ambulatory adult population, is at $33.71 billion in 2013 US dollars. This figure, representing more than 69% of the estimated societal cost, is obtained by multiplying the per-capita incremental cost of anxiety ($1657.52) by the national estimated prevalence of anxiety disorders in MEPS (20.34 million persons).

Previous estimates of direct medical costs of anxiety disorders, which were obtained in the late 90’s, range from $28.73 billion to $98.26 billion in 2013 US dollars.5,10 Even though our estimate falls within this range, it is not quite comparable with the previous findings. The type of methodology used to estimate the costs has a significant impact on results. DuPont et al.5 estimated the medical expenditures as the product of volume of services and unit prices or charges, and Greenberg et al.10 estimates were derived from a two-step multivariate regression approach, while we used the incremental cost approach. In addition, their study population was not limited to the U.S. ambulatory adult population, as ours was. Also, they did not include all diagnoses of anxiety disorders in their analysis and considered different cost categories in estimating cost of illness. For instance, DuPont et al.5 used charges instead of costs, did not include costs due to PTSD, and almost 70% of their cost estimate ($19.9 billion) was due to costs for institutionalized population, i.e. those who reside in nursing homes and specialty mental health organizations. Greenburg et al.10 estimated cost of anxiety for individuals aged 15 to 54. They found that more than half of the costs of these disorders ($53 billion) were attributable to non-psychiatric direct medical expenditures. Interpreting this result, Greenburg et al. explained that “estimation of this component was based on results from a single staff–model HMO that may not be fully generalizable to the entire population.”10(p. 431)

Cost by Category of Healthcare Services

With respect to categories of direct medical cost, inpatient visits (35.00%), prescription medications (32.78%), and office-based medical provider visits (22.34%) together accounted for almost 93% of the overall incremental costs associated with anxiety disorders. Emergency room cost, representing almost 3% of overall medical costs, was also slightly higher for sufferers of anxiety. These findings are consistent with the known healthcare utilization pattern and treatment seeking behavior of individuals with anxiety disorders. Marciniak, Lage, Landbloom, Dunayevich, and Bowman15 estimated the medical and productivity cost of anxiety disorders using data from a large employer database. Their results showed that employees with anxiety disorders had

73

higher inpatient hospital costs as well as higher prescription medications, medical provider visits, and emergency care costs.

The high cost of office-based medical provider visits and prescription medications can also be explained through the treatment seeking behavior of individuals with anxiety disorders. Treatment options for anxiety disorders include pharmacotherapy, psychotherapy, or combination of both. With the recent advances in psychotherapy techniques and pharmacotherapy products, such as Cognitive-Behavioral Therapy (CBT) and Selective Serotonin Reuptake Inhibitors (SSRIs) proven to be very effective in treating anxiety disorders, treatment utilization for these conditions has significantly increased throughout the time.87,88 As such, office-based medical provider visits and prescription medications are expected to account for the majority of overall cost of illness associated with anxiety disorders.

. To effectively reduce the cost of anxiety disorders, more attention needs to be geared towards these three categories of care. For instance, further studies should investigate the underlying reasons for high hospitalizations in patients with anxiety disorders, and examine whether these hospitalizations are potentially preventable.

Cost for Different Sub-populations

Knowing which sub-population incurs higher costs, policymakers and clinicians will be able to develop tailored disease management programs, by considering the specific characteristics of the target sub-populations.

Our results showed that the following sub-populations accounted for the majority of the overall direct medical costs associated with anxiety disorders in their category: Females ($2137.98), individuals 65 years and older ($2426.40), White non-Hispanics ($1879.31), previously married individuals ($2305.23), those with high levels of education (i.e. master/PhD) ($2463.31), middle-income earners ($2048.08), those who reside in the Midwest ($2338.59), non-MSA residents ($2485.64), and dual eligible ($6455.84). The only sub-populations, in which having anxiety disorder(s) was not associated with higher medical cost, were Hispanics, individuals with less than a high school diploma/GED, the uninsured, and those who were covered only by Medicaid, or other public insurances (such as Tricare).

Higher costs for females and White non-Hispanics can be explained through the higher prevalence of anxiety disorders among these sub-populations.30 With respect to insurance coverage, dual eligible followed by Medicare enrollees and private insurance holders had the highest portion of medical costs. These plans usually offer more generous benefits than Medicaid and other forms of public insurance. As such, it is expected for their members to have higher utilization and cost for services such as psychotherapy session, which are either not covered or limitedly covered by other plan types. The life- time prevalence of anxiety disorders is the lowest for people 65 years and older; yet we found that this group of individuals accounted for the majority of cost. This can be

74

explained through the correlation between age and Medicare insurance coverage. Patients with middle income, as well as those with high educational level had high anxiety-related costs. This observation can be justified assuming that people with higher socioeconomic status are more aware of the importance of treatment seeking for their diseases.

As it was mentioned above, anxiety disorders were not associated with an increase in healthcare cost among Hispanics. Also, anxiety related costs for Black non- Hispanics, even though statistically significant, were less than costs for White non- Hispanics. One explanation for these observations could be the lower life-time prevalence of anxiety disorders among Hispanics and Black non-Hispanics, in comparison with White non-Hispanics.30 However, future research needs to be done to examine any potential barrier in receiving the treatment for anxiety disorders among minorities.

To our knowledge, there is no study in the literature examining cost of anxiety among different sub-populations. Murciniak et al.18 used MarketScan Databases to examine how medical conditions and demographic characteristics affect the costs of treating anxiety patients. The only demographic characteristics included in their analyses were gender, age, and insurance coverage. Similar to our findings, they concluded that females, older individuals, and those who have more comprehensive insurance coverage incurred higher costs.18

We broke down anxiety costs for Blacks and Whites, by sex, gender and geographic region. Our findings (i.e. considerable variation in incremental costs incurred by different sub-populations), demonstrate why it is important to calculate values for sub- groups; and highlight the need for future studies to figure out the underlying causes of such variations. Of particular interest is the much larger costs for Black non-Hispanics in the Northeast ($3899.40) and for Black non-Hispanic males aged 45 and older ($3932.22). These findings deserve additional study to determine the reasons for such higher costs in theses sub-populations.

Indirect Costs

Based on the prevalence of self-reported anxiety disorders in MEPS and suicide data from NVSS, the total annual indirect cost attributable to anxiety disorders was estimated at $15.06 billion in 2013 US dollars ($12.72 for morbidity $2.34 for mortality cost). This figure represents almost 31% of the estimated societal cost of anxiety disorders.

Like direct medical cost, the estimated morbidity cost is also based on the prevalence of self-reported anxiety disorders in MEPS. So, there is a possibility of under- estimation of morbidity cost in this study (due to potential under-reporting of anxiety disorders in MEPS).

There are only two studies that provided national estimates of indirect costs for anxiety disorders. Greenberg et al.10 estimations of morbidity and mortality costs are

75

$7.44 billion and $2.12 billion (in 2013 US dollars) respectively. Even though their analysis technique for mortality cost is pretty similar to ours, their estimation of morbidity cost is not quite comparable with our results, because:

x They estimated productivity loss due to anxiety disorders only for the employed individuals, while we considered both employed and unemployed individuals in the analysis.

x For employed individuals, they defined morbidity cost as the cost due to absenteeism, plus reduced productivity while at work. The latter represented more than 87% of their estimated morbidity cost. However, we did not include this component (reduced productivity while at work) in our analysis.

x To get the number of missed work days due to anxiety disorders, we used regression analysis while they applied a 40% impairment rate to the total number of work cutbacks.

DuPont et al.,5 also provided estimates of morbidity and mortality cost at $59.83 billion and $2.30 billion (in 2013 US dollars), respectively. In calculating the mortality cost, they adopted the same methodology as ours, but their estimation of morbidity cost was based on impairment rate, and thus is not comparable with our findings.

Societal Cost

The societal cost of anxiety disorders were estimated at 48.72 billion in 2013 US dollars. Figure 5-1 shows the proportion of each cost component from the societal cost of anxiety disorders for the ambulatory adult population of the United States.

This study used nationally representative databases along with a robust statistical analysis technique to provide the most comprehensive and recent estimates of societal cost of anxiety disorders among adults in the U.S. The current study demonstrates conclusively that anxiety disorders, with the annual societal cost of $48.72 billion in 2013 US dollars, absorb a significant portion of US healthcare resources and should be prioritized by policymakers and healthcare providers who aim to reduce downstream costs of mental disorders.

Almost 70% of societal cost of anxiety disorders was due to direct medical costs. We analyzed this category further by looking at the distribution of costs in different health delivery settings, and amongst different sub-populations.

Our findings may also influence policy under the new Affordable Care Act (ACA). The ACA, signed into law on March 2010, aims to: expand coverage for all Americans, enhance the quality of care, and lower healthcare costs.89 In this regard, economic evaluations, such as cost of illness studies, can serve as an important tool in creating a healthcare system with lower costs and higher quality of care. Cost of illness

76

Figure 5-1. Percentage of each cost component from the societal cost.

studies are also useful in being able track healthcare costs over time to see if the increase access to healthcare and the cost-reductions in the ACA are having an effect. Cost of illness studies enable policymakers to identify medical conditions that absorb a significant portion of the US healthcare budget. These costly medical conditions may be investigated further to realize if resources are being distributed inefficiently, or if there is a need to invest in new, cost-effective treatment options. All these will lead to prioritizing scarce healthcare resources in a more efficient way, which eventually may lower the cost of care. However, the ACA lacks a mechanism to directly use economic evaluations, to reach these goals. As such, new healthcare policies are needed to support research that will help prioritize allocation of scarce healthcare resources.

Limitations

This study has several limitations. Most importantly, our findings might have underestimated the direct medical expenditures, as well as indirect cost aspect of morbidity, attributable to anxiety disorders. As it was discussed earlier, individuals with anxiety disorders were identified as those who reported being diagnosed with this condition. So, prevalence of anxiety disorders might be under-reported in MEPS. As such, our findings should be interpreted with caution. Results from this study, due to potential under-representation of anxiety disorders in MEPS, should be interpreted as a conservative estimate of the societal cost of these conditions in the ambulatory adult population of the U.S. If we assume that health seeking behavior and healthcare utilization of individuals with anxiety disorders, who didn’t report their condition, is not systematically different from other individuals in the study population, then we can apply the estimated per-person incremental medical cost ($1657.52) to the NCS-R’s estimate of the 12-month prevalence of anxiety disorders (i.e. 18.1% of the adult population which is equal to 42.13 million persons); and it gives us the total direct medical cost of anxiety

77

disorders at $69.84 billion in 2013 US dollars. Similarly, assuming that productivity loss of sufferers who reported their anxiety is consistent with the rest of the population, the estimated morbidity cost would be $25.36 billion.

Second, some covariates such as history and severity of illness were not included in the analysis of direct medical cost, due to unavailability of this information in MEPS. Third, we studied the societal cost of anxiety disorders in the ambulatory adult population of the U.S. A more comprehensive study including all age groups, as well as patients in assisted living or nursing home facilities, in analysis would provide a more precise estimate of the economic burden of these conditions. Fourth, since only the first three digits of ICD-9 codes are shown in MEPS public use files, we couldn’t unbundle the umbrella category of anxiety disorders and report costs by each diagnosis of these conditions. Finally, in order to find the number of deaths due to anxiety disorders, we applied a 10% rate to the total number of suicides. Even though we have adopted this rate from previous relevant research, it may not be a very accurate estimate for anxiety- induced suicides. Unfortunately, the national number of suicides for each specific diagnosis of mental illnesses is not still available.

78

LIST OF REFERENCES

1. National Health Expenditures 2011 Highlights. [Centers for Medicare & Medicaid Services]. 2012; http://www.cms.gov/Research-Statistics-Data-and- Systems/Statistics-Trends-and- Reports/NationalHealthExpendData/downloads/highlights.pdf. Accessed 01/24/2013.

2. OECD Health Data 2012 How Does the United States Compare. [Organisation for Economic Co-operation and Development]. 2012; http://www.oecd.org/unitedstates/BriefingNoteUSA2012.pdf. Accessed 01/24/2013.

3. Joel E, Segel BA. Cost-of-Illness Studies—A Primer. RTI-UNC Center of Excellence in Health Promotion Economics;2006.

4. Rice DP. Cost of illness studies: what is good about them? Inj Prev. Sep 2000;6(3):177-179.

5. DuPont RL, Rice DP, Miller LS, Shiraki SS, Rowland CR, Harwood HJ. Economic costs of anxiety disorders. Anxiety. 1996;2(4):167-172.

6. Anxiety Disorders- Common Characteristics. [AllPsych Online]. http://allpsych.com/disorders/anxiety/index.html. Accessed 09/01/2012.

7. Diagnostic and Statistical Manual of Mental Disorders (4th ed., text rev.). Washington, DC: American Psychiatric Association; 2000.

8. Anxiety Disorders. Washington, DC: U.S. DEPARTMENT OF HEALTH AND HUMAN SERVICES; 2009.

9. Kessler RC, Chiu WT, Demler O, Merikangas KR, Walters EE. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the National Comorbidity Survey Replication. Archives of general psychiatry. Jun 2005;62(6):617-627.

10. Greenberg PE, Sisitsky T, Kessler RC, et al. The economic burden of anxiety disorders in the 1990s. Journal of Clinical Psychiatry. Jul 1999;60(7):427-435.

11. Goetzel RZ, Hawkins K, Ozminkowski RJ, Wang S. The health and productivity cost burden of the "top 10" physical and mental health conditions affecting six large U.S. employers in 1999. Journal of occupational and environmental medicine / American College of Occupational and Environmental Medicine. Jan 2003;45(1):5-14.

79

12. Andlin-Sobocki P, Jonsson B, Wittchen HU, Olesen J. Cost of disorders of the brain in Europe. European Journal of Neurology. Jun 2005;12 Suppl 1:1-27.

13. Andlin-Sobocki P, Wittchen HU. Cost of anxiety disorders in Europe. European Journal of Neurology. Jun 2005;12 Suppl 1:39-44.

14. Smit F, Cuijpers P, Oostenbrink J, Batelaan N, de Graaf R, Beekman A. Costs of nine common mental disorders: implications for curative and preventive psychiatry. The journal of mental health policy and economics. Dec 2006;9(4):193-200.

15. Marciniak M, Lage MJ, Landbloom RP, Dunayevich E, Bowman L. Medical and productivity costs of anxiety disorders: case control study. Depression and anxiety. 2004;19(2):112-120.

16. Leon AC, Olfson M, Portera L. Service utilization and expenditures for the treatment of panic disorder. General hospital psychiatry. Mar 1997;19(2):82-88.

17. Rees CS, Richards JC, Smith LM. Medical utilisation and costs in panic disorder: a comparison with social phobia. Journal of Anxiety Disorders. Sep-Oct 1998;12(5):421-435.

18. Marciniak MD, Lage MJ, Dunayevich E, et al. The cost of treating anxiety: the medical and demographic correlates that impact total medical costs. Depression and anxiety. 2005;21(4):178-184.

19. Panzer PE, Regan TS, Chiao E, Sarnes MW. Implications of an SSRI generic step therapy pharmacy benefit design: an economic model in anxiety disorders. Am J Manag Care. Oct 2005;11(12 Suppl):S370-379.

20. McLaughlin TP, Khandker RK, Kruzikas DT, Tummala R. Overlap of anxiety and depression in a managed care population: Prevalence and association with resource utilization. Journal of Clinical Psychiatry. Aug 2006;67(8):1187-1193.

21. Stein MB, Cantrell CR, Sokol MC, Eaddy MT, Shah MB. Antidepressant adherence and medical resource use among managed care patients with anxiety disorders. Psychiatric services (Washington, D.C.). May 2006;57(5):673-680.

22. Olfson M, Gameroff MJ. Generalized anxiety disorder, somatic pain and health care costs. General hospital psychiatry. Jul-Aug 2007;29(4):310-316.

23. Roberge P, Marchand A, Reinharz D, et al. Healthcare utilization following cognitive-behavioral treatment for panic disorder with agoraphobia. Cogn Behav Ther. 2005;34(2):79-88.

80

24. Siegel L, Jones WC, Wilson JO. Economic and life consequences experienced by a group of individuals with panic disorder. Journal of Anxiety Disorders. // 1990;4(3):201-211.

25. Souetre E, Lozet H, Cimarosti I, et al. Cost of anxiety disorders: impact of comorbidity. Journal of psychosomatic research. 1994;38 Suppl 1:151-160.

26. Salvador-Carulla L, Segui J, Fernandez-Cano P, Canet J. Costs and offset effect in panic disorders. British Journal of Psychiatry - Supplement. Apr 1995(27):23-28.

27. Patel A, Knapp M, Henderson J, Baldwin D. The economic consequences of social phobia. Journal of affective disorders. Apr 2002;68(2-3):221-233.

28. Batelaan N, Smit F, de Graaf R, van Balkom A, Vollebergh W, Beekman A. Economic costs of full-blown and subthreshold panic disorder. Journal of Affective Disorders. Dec 2007;104(1-3):127-136.

29. Acarturk C, Smit F, de Graaf R, van Straten A, Ten Have M, Cuijpers P. Economic costs of social phobia: a population-based study. Journal of affective disorders. Jun 2009;115(3):421-429.

30. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. Jun 2005;62(6):593-602.

31. Kirschstein R. Disease-Specific Estimates of Direct and Indirect Costs of Illness and NIH Support: Fiscal Year 2000 Update. 2005; http://ospp.od.nih.gov/ecostudies/COIreportweb.htm.

32. Hodgson TA, Meiners MR. Cost-of-illness methodology: a guide to current practices and procedures. Milbank Mem Fund Q Health Soc. Summer 1982;60(3):429-462.

33. MEPS HC-135I: Appendix to MEPS 2010 Event Files HC-135A - HC-135H Rockville, MD: Agency for Healthcare Research and Quality-Center for Financing, Access, and Cost Trends; 2012.

34. Rice DP. Estimating the cost of illness. Am J Public Health Nations Health. Mar 1967;57(3):424-440.

35. Koopmanschap MA, van Ineveld BM. Towards a new approach for estimating indirect costs of disease. Soc Sci Med. May 1992;34(9):1005-1010.

36. Koopmanschap MA, Rutten FF, van Ineveld BM, van Roijen L. The friction cost method for measuring indirect costs of disease. J Health Econ. Jun 1995;14(2):171-189.

81

37. Johannesson M, Karlsson G. The friction cost method: a comment. J Health Econ. Apr 1997;16(2):249-255; discussion 257-249.

38. Encyclopedia of Public Health. Vol 1. 1 ed: Springer; 2008.

39. Chapin CV. The value of human life. American journal of public health (New York, N.Y. : 1912). Feb 1913;3(2):101-105.

40. Mushkin SJ, Collings FD. Economic costs of disease and injury. Public health reports (Washington, D.C. : 1974). Sep 1959;74:795-809.

41. Fein R. Economics of Public Health. New York: Basic Books; 1958.

42. Drummond M. Cost-of-illness studies: a major headache? PharmacoEconomics. Jul 1992;2(1):1-4.

43. Edlund MJ, Swann AC. The economic and social costs of panic disorder. Hosp Community Psychiatry. Dec 1987;38(12):1277-1279, 1288.

44. Konnopka A, Leichsenring F, Leibing E, Konig HH. Cost-of-illness studies and cost-effectiveness analyses in anxiety disorders: a systematic review. J Affect Disord. Apr 2009;114(1-3):14-31.

45. Survey Background. 2009. http://meps.ahrq.gov/mepsweb/about_meps/survey_back.jsp.

46. MEPS HC-138: 2010 Full Year Consolidated Data File. 2012; http://meps.ahrq.gov/mepsweb/data_stats/download_data/pufs/h138/h138doc.sht ml#2581UnitedStates. Accessed 4/01/2013.

47. Machlin SR, Zodet MW, Nixon JA. Estimates of medical expenditures from the Medical Expenditure Panel Survey: Gains in precision from combining consecutive years of data. Paper presented at: Joint Statistical Meetings - Section on Survey Research Methods2003; Alexandria, VA.

48. MEPS HC-036BRR: 1996-2009 Replicates for Variance Estimation File. Rockville, MD: Agency for Healthcare Research and Quality-Center for Financing, Access, and Cost Trends;2011.

49. MEPS HC-137: 2010 Medical Conditions. 2012; http://meps.ahrq.gov/mepsweb/data_stats/download_data/pufs/h137/h137doc.sht ml. Accessed 4/01/2013.

82

50. Shirneshan E, Bailey J, Relyea G, Franklin BE, Solomon DK, Brown LM. Incremental direct medical expenditures associated with anxiety disorders for the U.S. adult population: Evidence from the Medical Expenditure Panel Survey. Journal of Anxiety Disorders. 10// 2013;27(7):720-727.

51. BLS Information. [Bureau of Labor Statistics]. http://www.bls.gov/dolfaq/bls_ques26.htm. Accessed 10/01/2012.

52. Affordable Care Act. 2013; http://www.medicaid.gov/affordablecareact/affordable-care-act.html. Accessed 08/21/2013.

53. Labor Force Statistics from the Current Population Survey. Bareau of Labor Statistics. http://bls.gov/data/. Accessed 11/05/2012.

54. CPI Detailed Report Data for March 2013. Washington, DC U.S. Bureau of Labor Statistics, Division of Consumer Prices and Price Indexes; 05/10/2013 2013.

55. About NCHS. [Center for Disease Control and Prevention]. http://www.cdc.gov/nchs/about/policy/quality.htm. Accessed 11/01/2012.

56. Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 5-year age groups, by Hispanic origin, race for non-Hispanic population and sex: United States, 2010. Mortality Tables 2012; http://www.cdc.gov/nchs/nvss/mortality_tables.htm. Accessed 11/25/2012.

57. Deaths, percent of total deaths, and death rates for the 15 leading causes of death in 10-year age groups, by Hispanic origin, race for non-Hispanic population and sex: United States, 2010. Mortality Tables 2012; http://www.cdc.gov/nchs/nvss/mortality_tables.htm. Accessed 11/25/2012.

58. Deaths, percent of total deaths and rank order for 113 selected causes of death and Enterocolitis due to Clostridium difficile, by Hispanic origin, race for non- Hispanic origin and sex, United States, 2010. Mortality Tables 2012; http://www.cdc.gov/nchs/nvss/mortality_tables.htm. Accessed 11/25/2012.

59. Murphy SL, Xu JQ, Kochanek KD. Deaths: Final Data for 2010. National Vital Statistics Reports. Hyattsville, MD: National Center for Health Statistics;2013.

60. Balu S, Thomas J, 3rd. Incremental expenditure of treating hypertension in the United States. Am J Hypertens. Aug 2006;19(8):810-816; discussion 817.

61. Bhattacharyya N. Incremental healthcare utilization and expenditures for allergic rhinitis in the United States. The Laryngoscope. 2011;121(9):1830-1833.

83

62. Kamble S, Bharmal M. Incremental direct expenditure of treating asthma in the United States. Journal of Asthma. Feb 2009;46(1):73-80.

63. Kawatkar AA, Jacobsen SJ, Levy GD, Medhekar SS, Venkatasubramaniam KV, Herrinton LJ. Direct medical expenditure associated with rheumatoid arthritis in a nationally representative sample from the Medical Expenditure Panel Survey. Arthritis care & research. Nov 2012;64(11):1649-1656.

64. Luo X, Pietrobon R, Sun SX, Liu GG, Hey L. Estimates and patterns of direct health care expenditures among individuals with back pain in the United States. Spine. Jan 1 2004;29(1):79-86.

65. Olin G, Machlin SR, Rhoades J. Estimating the cost of illness: The case of diabetes. 2008.

66. Raval AD, Sambamoorthi U. Incremental healthcare expenditures associated with thyroid disorders among individuals with diabetes. Journal of thyroid research. 2012;2012.

67. D'Hoore W, Bouckaert A, Tilquin C. Practical considerations on the use of the Charlson comorbidity index with administrative data bases. Journal of clinical epidemiology. Dec 1996;49(12):1429-1433.

68. Buntin MB, Zaslavsky AM. Too much ado about two-part models and transformation?: Comparing methods of modeling Medicare expenditures. Journal of Health Economics. 5// 2004;23(3):525-542.

69. Breusch TS, Pagan AR. A Simple Test for Heteroscedasticity and Random Coefficient Variation. Econometrica. 1979;47(5):1287-1294

70. Park R. Estimation with Heteroscedastic Error Terms. Econometrica. 1966;34(4):888.

71. Industries at a Glance. [Bureau of Labor Statistics]. http://www.bls.gov/iag/tgs/iag814.htm. Accessed 11/01/2012.

72. Akazawa M, Sindelar JL, Paltiel AD. Economic costs of influenza-related work absenteeism. Value in Health. 2003;6(2):107-115.

73. Rice DP, Kelman S, Miller LS. Estimates of economic costs of alcohol and drug abuse and mental illness, 1985 and 1988. Public health reports (Washington, D.C. : 1974). May-Jun 1991;106(3):280-292.

74. Employment Cost Index. Bureau of Labor Statistics. http://www.bls.gov/data/. Accessed 06/ 24/2013.

84

75. Siegel JE, Torrance GW, Russell LB, Luce BR, Weinstein MC, Gold MR. Guidelines for pharmacoeconomic studies. Recommendations from the panel on cost effectiveness in health and medicine. Panel on cost effectiveness in health and medicine. PharmacoEconomics. Feb 1997;11(2):159-168.

76. SAS Institute Inc. SAS/STAT® 9.3 User’s Guide. Cary. NC: SAS Institute Inc; 2011.

77. StataCorp. Stata Statistical Software: Release 12. College Station, TX: StataCorp. LP; 2011.

78. O’brien R. A Caution Regarding Rules of Thumb for Variance Inflation Factors. Qual Quant. 2007/10/01 2007;41(5):673-690.

79. Schoen C, Osborn R, Squires D, Doty MM, Pierson R, Applebaum S. How health insurance design affects access to care and costs, by income, in eleven countries. Health affairs (Project Hope). Dec 2010;29(12):2323-2334.

80. Buchmueller TC, Grumbach K, Kronick R, Kahn JG. Book Review: The Effect of Health Insurance on Medical Care Utilization and Implications for Insurance Expansion: A Review of the Literature. Medical Care Research and Review. February 1, 2005 2005;62(1):3-30.

81. Hurd MD, McGarry K. Medical insurance and the use of health care services by the elderly. Journal of Health Economics. 4// 1997;16(2):129-154.

82. Stanton MW, Rutherford MK. The high concentration of U.S. health care expenditures. Rockville, MD: Agency for Healthcare Research and Quality;2005. 06-0060.

83. Alemayehu B, Warner KE. The lifetime distribution of health care costs. Health services research. Jun 2004;39(3):627-642.

84. Kim MH, Johnston SS, Chu BC, Dalal MR, Schulman KL. Estimation of total incremental health care costs in patients with atrial fibrillation in the United States. Circulation. Cardiovascular quality and outcomes. May 2011;4(3):313- 320.

85. Yelin E, Trupin L, Cisternas M, Eisner M, Katz P, Blanc P. A national study of medical care expenditures for respiratory conditions. European Respiratory Journal. Mar 2002;19(3):414-421.

86. Yoon D, Frick KD, Carr DA, Austin JK. Economic impact of epilepsy in the United States. Epilepsia. Oct 2009;50(10):2186-2191.

85

87. Olfson M, Marcus SC, Wan GJ, Geissler EC. National trends in the outpatient treatment of anxiety disorders. Journal of Clinical Psychiatry. Sep 2004;65(9):1166-1173.

88. Treatment. Anxiety Disorder association of America 2013; http://www.adaa.org/finding-help/treatment. Accessed 06/11/2013.

89. Affordable Care Act. http://www.medicaid.gov/affordablecareact/affordable-care- act.html. Accessed 08/21/2013.

86

VITA

Elaheh Shirneshan was born in Iran in 1981. She finished her bachelor’s and master’s degree in Industrial Engineering in 2005 and 2009, respectively. In fall 2010, she was accepted at the University of Tennessee Health Science Center as a graduate student in the Ph.D. program (Major: Health Outcomes and Policy Research).She graduated from this program in December 2013.

87 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA)

COVID-19 is an emerging, rapidly evolving situation.

Get the latest information from CDC (coronavirus.gov) | NIH Resources | NIDA Resources

National Institutes of Health Order Publications En español

Search

Trends & Statistics /

 Trends & Statistics Overdose Death Rates

The U.S. government does not track death rates for every drug. However, the National Center for Health Statistics at the Centers for Disease Control and Prevention does collect information on many of the more commonly used drugs. The CDC also has a searchable database, called CDC Wonder.

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 1. National Drug Overdose Deaths—Number Among All Ages, by Gender, 1999-2018. More than 67,300 Americans died from drug-involved overdose in 2018, including illicit drugs and prescription opioids. The gure above is a bar and line graph showing the total number of U.S. drug overdose deaths involving any illicit or prescription opioid drug from 1999 to 2018. Drug overdose deaths rose from 38,329 in 2010 to 70,237 in 2017; followed by a signicant decrease in 2018 to 67,367 deaths. The bars are overlaid by lines showing the number of deaths by gender from 1999 to 2018 (Source: CDC WONDER). https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 1/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA)

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 2. National Drug Overdose Deaths by Specic Category—Number Among All Ages, 1999-2018. Overall, drug overdose deaths declined from 2017 to 2018 with 67,637 drug overdose deaths reported in 2018. Deaths involving other synthetic narcotics other than methadone (including fentanyl and fentanyl analogs) continued to rise with more than 31,335 overdose deaths reported in 2018. Those involving cocaine or psychostimulants with abuse potential (mostly methamphetamine) also continued to trend upward (Source: CDC WONDER).

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 3. National Drug Overdose Deaths Involving Any Opioid—Number Among All Ages, by Gender, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving any opioid from 1999 to 2018. Any opioid includes prescription opioids (and methadone), heroin and other synthetic narcotics (mainly fentanyl or fentanyl analogs). Opioid-involved overdose deaths rose from 21,088 in 2010 to 47,600 in 2017 and remained steady in 2018 with 46,802 deaths. The bars are overlaid by lines showing the number of deaths by gender from 1999 to 2018 (Source: CDC WONDER).

https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 2/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA)

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 4. National Overdose Deaths Involving Prescription Opioids—Number Among All Ages, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving prescriptions opioids (including methadone) from 1999 to 2018. Drug overdose deaths involving prescription opioids rose from 3,442 in 1999 to 17,029 in 2017. From 2017 to 2018, however, the number of deaths dropped to 14,975. The bars are overlaid by lines showing the number of deaths by gender from 1999 to 2018 (Source: CDC WONDER).

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 5. National Overdose Deaths Involving Heroin, by Other Synthetic Narcotic (Opioid) Involvement, Number Among All Ages, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving heroin from 1999 to 2018. Drug overdose deaths involving heroin rose from 1,960 in 1999 to 15,469 in 2016. Since 2016, the https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 3/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA) number of deaths has remained steady with 14,996 deaths reported in 2018. The bars are overlaid by lines showing the number of deaths involving heroin in combination with or without other synthetic narcotics (mainly fentanyl or fentanyl analogs) from 1999 to 2018. The number of deaths involving heroin in combination with synthetic narcotics has been increasing steadily since 2014 and shows that the increase in deaths involving heroin is driven by the use of fentanyl (Source: CDC WONDER).

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 6. National Drug Overdose Deaths Involving Psychostimulants With Abuse Potential (Including Methamphetamine), by Opioid Involvement, Number Among All Ages, 1999- 2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving psychostimulants with abuse potential from 1999 to 2018. Drug overdose deaths rose from 547 in 1999 to 12,676 in 2018. The bars are overlaid by lines showing the number of deaths involving psychostimulants with or without any opioid, and involving psychostimulants with or without other synthetic narcotics. The number of deaths involving psychostimulants has increased steadily since 2014 regardless of opioid involvement (Source: CDC WONDER).

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 4/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA) on CDC WONDER Online Database, released January, 2020

Figure 7. National Drug Overdose Deaths Involving Cocaine, by Opioid Involvement, Number Among All Ages, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving cocaine from 1999 to 2018. Drug overdose deaths involving cocaine rose from 3,822 in 1999 to 13,942 in 2017 and remained steady in 2018 with 14,666. The bars are overlaid by lines showing the number of deaths involving cocaine with or without any opioid, and cocaine and other synthetic narcotics. The number of deaths in combination with any opioid has been increasing steadily since 2014 and is mainly driven by the involvement of other synthetic narcotics (mainly fentanyl or fentanyl analogs) (Source: CDC WONDER).

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 8. National Drug Overdose Deaths Involving Benzodiazepines, by Opioid Involvement, Number Among All Ages, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving benzodiazepines from 1999 to 2018. Drug overdose deaths involving benzodiazepines rose from 1,135 in 1999 to 11,537 in 2017. Between 2017 and 2018, deaths declined to 10,724.The bars are overlaid by lines showing the number of deaths involving benzodiazepines with or without any opioid, and benzodiazepines and other synthetic narcotics. The number of deaths involving benzodiazepines in combination with other synthetic narcotics has been increasing steadily since 2014, while deaths involving benzodiazepines without any opioids has remained steady (Source: CDC WONDER).

https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 5/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA)

Source: Centers for Disease Control and Prevention, National Center for Health Statistics. Multiple Cause of Death 1999-2018 on CDC WONDER Online Database, released January, 2020

Figure 9. National Drug Overdose Deaths Involving Antidepressants, by Opioid Involvement–Number Among All Ages, 1999-2018. The gure above is a bar and line graph showing the total number of U.S. overdose deaths involving antidepressants from 1999 to 2018. Drug overdose deaths involving antidepressants has risen steadily from 1,749 in 1999 to 5,269 in 2017. In 2018, deaths remained steady with 5,064. The bars are overlaid by lines showing the number of deaths involving antidepressants with or without any opioid, and antidepressants and other synthetic narcotics (Source: CDC WONDER).

The gures above are bar charts showing the number of U.S. overdose deaths involving select prescription and illicit drugs from 1999 through 2018. The bars are overlaid by lines representing gender or concurrent opioid involvement. There were 67,367 drug-involved overdose deaths reported in the U.S. in 2018 (Figure 1); 67% of cases occurred among males (yellow line). Other synthetic narcotics or opioids (mainly fentanyl or fentanyl analogs) were the main driver of drug overdose deaths with a nearly 12-fold increase from 2012 to 2018 (Figure 2).

Drug overdose deaths involving any opioid―prescription opioids (including methadone), other synthetic opioids, and heroin―rose from 18,515 deaths in 2007 to 47,600 deaths in 2017; before declining to 46,802 in 2018. More than 68% of deaths occurred among males (Figure 3). From 2017 to 2018, the number of deaths involving prescription opioids declined to 14,975 (Figure 4).

Overdose deaths involving heroin decreased from 15,482 deaths in 2017 to 14,996 in 2018 (Figure 5).

From 2015 to 2018, the number of deaths involving psychostimulants (mainly methamphetamine, Figure 6) or cocaine (Figure 7) have risen signicantly to a respective 12,676 and 14,666 deaths.

https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 6/7 12/27/2020 Overdose Death Rates | National Institute on Drug Abuse (NIDA) The nal two charts show the number of overdose deaths involving benzodiazepines (Figure 8) or antidepressants (Figure 9). Benzodiazepines were involved in 10,734 deaths in 2018—a decrease from the 11,537 deaths in 2017. These were driven by the combination of these prescription drugs with any opioid. Deaths involving antidepressants are also rising, although at a much slower rate than benzodiazepines. As is the case with benzodiazepines, deaths involving antidepressants are mainly driven by those also involving synthetic opioids.

Download presentation le -  National Drug Overdose Deaths 1999-2018 (Powerpoint, 215KB)

Download  Drug Overdoses data document (XLS, 177KB)

NIDA for Teens: Drug Overdoses in Youth

March 10, 2020

https://www.drugabuse.gov/drug-topics/trends-statistics/overdose-death-rates 7/7 12/10/2020 https://emedicine.medscape.com/article/813255-print

emedicine.medscape.com

Benzodiazepine Toxicity

Updated: Jan 23, 2020 Author: Chip Gresham, MD, FACEM; Chief Editor: Gil Z Shlamovitz, MD, FACEP

Overview

Practice Essentials

Oral benzodiazepine (BZD) overdoses, without co-ingestions, rarely result in significant morbidity (eg, aspiration pneumonia, rhabdomyolysis) or mortality. In mixed overdoses, they can potentiate the effect of alcohol or other sedative-hypnotics. Acute intravenous administration of BZDs is associated with greater degrees of respiratory depression.

Patients receiving prolonged parenteral administration of BZDs are at risk for propylene glycol poisoning (the diluent used in parenteral formulations of diazepam and lorazepam). Although rare, this may result in hypotension, cardiac dysrhythmias, lactic acidosis, seizures, or coma.

In long-term users who have developed dependence, cessation of BZDs can result in a withdrawal syndrome, with manifestations including anxiety, irritability, confusion, seizures, and sleep disorders.[1] Alprazolam withdrawal syndrome may be especially severe, with associated delirium, psychosis, and hyperadrenergic states.[2] Treatment of BZD withdrawal is typically with a long-acting BZD (eg, clonazepam), but sucessful use of antiseizure drugs (eg, valproate, carbamazepine) has also been reported.[2]

Signs and symptoms

Symptoms of BZD overdose may include the following:

Dizziness Confusion Drowsiness Blurred vision Unresponsiveness Anxiety Agitation

Findings on physical examination may include the following:

Nystagmus Hallucinations Slurred speech Ataxia Coma Hypotonia Weakness Altered mental status, impairment of cognition Amnesia Paradoxical agitation Respiratory depression Hypotension

See Presentation for more detail.

Diagnosis

https://emedicine.medscape.com/article/813255-print 1/8 12/10/2020 https://emedicine.medscape.com/article/813255-print Immunoassay screening techniques are performed most commonly. These tests typically detect BZDs that are metabolized to desmethyldiazepam or oxazepam; thus, a negative screening result does not rule out the presence of a BZD.

Tests and procedures to obtain depend on the presentation, as follows:

Arterial blood gas (ABG) assay, if respiratory depression is present ECG, to evaluate for co-ingestants, particularly cyclic antidepressants Chest radiograph, if respiratory compromise is present Pregnancy test, in women of childbearing age

In cases of intentional overdose, measure the following:

Serum electrolytes Glucose BUN Creatinine clearance Ethanol level Acetaminophen level

See Workup for more detail.

Management

As with any overdose, the first step is an assessment of the patient's airway, breathing, and circulation, and these should be addressed rapidly as needed. In any patient with an altered mental status, a blood glucose level should be obtained immediately. The cornerstone of treatment in BZD overdoses is good supportive care and monitoring. Single-dose activated charcoal is not routinely recommended, as the risks far outweigh the benefit; BZD overdoses are very rarely fatal, and the resulting altered mental status greatly increases the risk of aspiration following oral charcoal dose.[3]

Flumazenil (Romazicon) is a specific antidote for BZD poisoning, but its use in acute BZD overdose is controversial and its risks usually outweigh any possible benefit.[4] In long-term BZD users, flumazenil may precipitate withdrawal and seizures; in patients taking BZDs for a medical condition, flumenazil may result in exacerbation of the condition. The ideal indication for flumazenil is isolated iatrogenic (eg, during conscious sedation) BZD overdose in BZD-naive patients.

See Treatment and Medication for more detail.

Background

Benzodiazepine (BZD) toxicity may result from overdose or from abuse. Since their introduction in 1960, BZDs have come to be widely used for a variety of indications, including seizures, anxiety, alcohol withdrawal, insomnia, drug-associated agitation, and muscle spasm. In addition, BZDs are used as preanesthetic agents, and are frequently combined with other medications for procedural sedation. BZD overdose often occurs in association with other substances.

For patient education information, see Benzodiazepine Abuse, First Aid for Poisoning in Children, and Child Safety Proofing.

Pathophysiology

Benzodiazepines (BZDs) act by potentiating the activity of gamma-aminobutyric acid (GABA), which is the major inhibitory neurotransmitter in the central nervous system (CNS). BZDs bind to a specific receptor on the GABA A receptor complex and thereby facilitate the binding of GABA to its specific receptor site. BZD binding causes increased frequency of opening of the chloride channel complexed with the GABA A receptor. Chloride channel opening results in membrane hyperpolarization, which inhibits cellular excitation.

Enhanced GABA neurotransmission results in sedation, striated muscle relaxation, anxiolysis, and anticonvulsant effects. Stimulation of peripheral nervous system (PNS) GABA receptors may cause decreased cardiac contractility and vasodilation. These changes could have the potential to alter tissue perfusion.

https://emedicine.medscape.com/article/813255-print 2/8 12/10/2020 https://emedicine.medscape.com/article/813255-print The rate of BZD onset of action is determined by its ability to cross the blood-brain barrier. The relatively lipophilic BZDs (eg, diazepam) usually have a faster onset of effect than the relatively water-soluble BZDs (eg, lorazepam). BZD effects can be potentiated when ethanol is present as a coingestant. Peak blood concentrations of most agents occur within 1-3 hours.

After a single dose, the lipophilic agents can have a shorter duration of action (shorter CNS effect) than water-soluble agents because rapid redistribution from the CNS to peripheral sites (eg, adipose tissue); thus, lorazepam has a longer CNS duration of action than diazepam. However, diazepam metabolizes to active intermediates with prolonged half-lives, which extend its therapeutic effects.

BZDs are metabolized predominantly in the liver by oxidation and/or conjugation. Most BZDs are broken down into pharmacologically active metabolites, which may have longer half-lives than the parent compounds.

Epidemiology

According to the Drug Abuse Warning Network (DAWN) of the Substance Abuse and Mental Health Services Administration (SAMHSA), total emergency department (ED) visits for nonmedical use of benzodiazepines (BZDs) rose 149% from 2004 to 2011. However, no short-term increases were noted between 2009 and 2011. Records did not always specify the BZD involved, but alprazolam was indicated in about a third of these ED visits, and in approximately a third of BZD-related suicide attempts.[5]

DAWN was discontinued in 2011. SAMHSA plans to combine DAWN into a new data collection, the Emergency Department Surveillance System (SEDSS).[6]

From 1996 to 2013, the percentage of US adults filling a BZD prescription increased 67%, from 8.1 million to 13.5 million, and the median cumulative quantity of BZDs filled over the year increased by 140%. The overdose death rate increased from 0.58 to 3.07 per 100,000 adults, with a plateau seen after 2010, although deaths continued to increase in the elderly and in blacks and Hispanics. In 2013, BZDs accounted for approximately 31% of fatal overdoses involving prescription drugs in the US. However, the danger of BZDs may be obscured by the fact that an estimated 75% of deaths involving benzodiazepines also involve an opioid.[7]

In 2018, a total of 22,995 single-substance exposures to BZDs were reported to US poison control centers. Of these, 469 (2%) resulted in major toxicity and 19 resulted in death.[8] Inclusion of cases involving BZDs in combination with alcohol or other drugs yields much higher numbers. DAWN reported that BZDs were involved in 123,572 of the 606,653 ED visits in 2011 that involved drugs and alcohol taken together (20.4%).[5]

The most reported cases of BZD toxicity are in persons older than 19 years.[8] The elderly and the very young are more susceptible to the CNS depressant effects of BZDs than people in other age groups.

Prognosis

Oral benzodiazepine (BZD) overdoses, without co-ingestions, rarely result in significant morbidity (eg, aspiration pneumonia, rhabdomyolysis) or mortality, although in mixed overdoses they can potentiate the effect of alcohol or other sedative-hypnotics.

Benzodiazepines are commonly misused by individuals with opioid dependence, with prevalence rates of 45–70% for patients in opioid maintenance treatment.[9] Although BZDs taken alone are relatively safe in overdose, the combination of BZDs and opioid analgesics can produce significant respiratory depression. In particular, the combination of alprazolam with opioids may be fatal. Analysis of a West Virginia forensic database showed that alprazolam contributed to 17% of drug-related deaths; at least one other drug, typically an opioid, was identified in 97.5% of the alprazolam cases, with concurrent BZDs also found.[10]

Overdose of ultrashort-acting BZDs (eg, triazolam [Halcion]) is also more likely to result in apnea and death than overdose with longer-acting BZDs. Of individual BZDs, alprazolam (Xanax) is relatively more toxic than others in overdose.[11] Deaths attributed to BZDs increased fivefold from 1999 to 2009. During 2003 to 2009, death rates from alprazolam increased 233.8%; alprazolam was second only to oxycodone among prescription drugs with the highest increase in death rates.[12]

Similarly, an Australian study reported that alprazolam-positive cases of sudden or unnatural death increased from three cases in 1997 to 86 cases in 2012. The increase was driven mostly by accidental toxicity in people with known drug and alcohol problems. Drugs other than alprazolam and its metabolites were present in 94.9% of cases. The most commonly detected drugs, in order of decreasing frequency, were opioids, other BZDs, and alcohol.[13] https://emedicine.medscape.com/article/813255-print 3/8 12/10/2020 https://emedicine.medscape.com/article/813255-print

Presentation

History

Information to elicit when taking the history in cases of possible benzodiazepine (BZD) overdose includes the following:

Time of ingestion Dose Co-ingestants, if any Whether the overdose was accidental or intentional In patients prescribed BZDs, the duration of BZD use

Symptoms of BZD overdose may include the following:

Dizziness Confusion Drowsiness Blurred vision Unresponsiveness Anxiety Agitation

Physical Examination

Focus the physical examination on the patient's vital signs and cardiorespiratory and neurologic function. "Classic" isolated benzodiazepine overdose presents as coma with normal vital signs.

Findings on physical examination may include the following:

Nystagmus Hallucinations Slurred speech Ataxia Coma Hypotonia Weakness Altered mental status, impairment of cognition Amnesia Paradoxical agitation Respiratory depression Hypotension

DDx

Differential Diagnoses

Acute Hypoglycemia

Alcohol Toxicity https://emedicine.medscape.com/article/813255-print 4/8 12/10/2020 https://emedicine.medscape.com/article/813255-print Antidepressant Toxicity

Encephalitis

Hypernatremia in Emergency Medicine

Hyponatremia in Emergency Medicine

Neuroleptic Agent Toxicity

Sedative-Hypnotic Toxicity

Stroke, Ischemic

Toxicity, Antihistamine

Workup

Workup

Approach Considerations

Overall, the laboratory detection of benzodiazepines (BZDs) depends upon the screening method used. Immunoassay screening techniques are performed most commonly and typically detect BZDs that are metabolized to desmethyldiazepam or oxazepam; thus, a negative screening result does not rule out the presence of a BZD. Qualitative screening of urine or blood may be performed but rarely influences treatment decisions and has no impact on immediate clinical care.

Tests and procedures to obtain depend on the presentation, as follows:

Arterial blood gas (ABG) assay, if respiratory depression is present ECG, to evaluate for co-ingestants, particularly cyclic antidepressants Chest radiograph, if respiratory compromise is present Pregnancy test, in women of childbearing age

In cases of intentional overdose, measure the following:

Serum electrolytes Glucose BUN Creatinine clearance Ethanol level Acetaminophen level

Treatment

Approach Considerations

As with any overdose, the first step is to assess the patient's airway, breathing, and circulation and to address those rapidly as needed. The cornerstone of treatment in benzodiazepine (BZD) overdoses is good supportive care and monitoring.

Single-dose activated charcoal is not routinely recommended, as the risks far outweigh the benefit. BZD are very rarely fatal in overdoses, and the altered mental status from BZD overdose greatly increases the risk of aspiration following oral charcoal dosing.[3]

Flumazenil (Romazicon) is a specific antidote for BZDs, but its use in acute BZD overdose is controversial and its risks usually outweigh any possible benefit.[4] In long-term BZD users, flumazenil may precipitate withdrawal and seizures; in patients taking BZDs for a medical condition, flumenazil may result in exacerbation of the condition. https://emedicine.medscape.com/article/813255-print 5/8 12/10/2020 https://emedicine.medscape.com/article/813255-print Disposition of patients is as follows:

Patients may be discharged if they remain asymptomatic at least 6 hours post ingestion. Patients with mild toxicity may be observed in the emergency department until they recover. Patients with intentional overdoses require psychiatric evaluation before discharge. Admit patients with hemodynamic instability, coma, or respiratory depression to the intensive care unit (ICU). Respiratory depression may be treated with assisted ventilation.

The American Psychiatric Association and the National Institute of Clinical Excellence have treatment and diagnostic guidelines available for cases of substance abuse and self-harm.[14, 15]

Prehospital Care

Prehospital care for patients who have overdosed on benzodiazepines (BZDs) includes the following:

Cardiac monitoring

Supplemental oxygen and airway support

Intravenous (IV) access

Rapid glucose determination (finger stick) and administration of D50 if necessary

Naloxone can be administered at a very low dose (0.05 mg with a gradual increase if needed) if the diagnosis is unclear and an opioid co-ingestion is suspected (eg, if the patient has severe respiratory depression).

An important caveat is that although the administration of 0.4 mg of naloxone will reverse respiratory depression in most patients with opioid overdoses, it will also cause severe withdrawal symptoms (nausea, vomiting) in those who are opioid dependent. This can result in aspiration of gastric contents in patients who are unable to protect their airway because of sedation from the BZD.

Flumazenil

Flumazenil is a competitive BZD receptor antagonist and is the only available specific antidote for BZDs. Its use in acute BZD is controversial, however, and its risks usually outweigh any benefit.[4] Common adverse events with flumazenil include agitation and gastrointestinal symptoms, while serious adverse events include supraventricular arrhythmia and convulsions.[16]

Flumazenil does not consistently reverse central respiratory depression due to BZDs, and over half the patients in a large multicenter study experienced re-sedation after use.[17]

In long-term BZD users, flumazenil may precipitate withdrawal and seizures; in patients taking BZDs for a medical condition, flumenazil may result in exacerbation of the condition.

In addition to those patients on long-term BZD use, flumazenil should not be used in any patient at an increased risk of having a seizure, including those with a seizure history, head injury, co-ingestion of BZD and tricyclic antidepressant or other proconvulsant, or even a possible ingestion of a proconvulsant.[18]

The ideal consideration for flumazenil use is for isolated iatrogenic BZD overdose in BZD-naive patients (eg, during conscious sedation of a BZD-naive patient).

Medication

Medication Summary https://emedicine.medscape.com/article/813255-print 6/8 12/10/2020 https://emedicine.medscape.com/article/813255-print Flumazenil is a selective competitive antagonist of the gamma-aminobutyric acid (GABA) receptor and is the only available specific antidote for benzodiazepine (BZD) toxicity; it will reverse the effects of BZDs but must be used with caution.

Antidotes, Other

Flumazenil

Flumazenil is a competitive BZD receptor antagonist and is the only available specific antidote for BZDs, although its use in acute BZD is controversial and its risks usually outweighs any benefit.

Flumazenil does not consitantly reverse central respiratory depression due to BZDs, and over half the patients in a large multicenter study experienced re-sedation after use.

In long-term BZD users, flumazenil may precipitate withdrawal and seizures; in patients taking BZDs for a medical condition, flumenazil may result in exacerbation of the condition.

In addition to those patients on long-term BZD use, flumazenil should not be used in any patient at an increased risk of having a seizure, including those with a seizure history, head injury, coingestion of BZD and a tricyclic antidepressant or other proconvulsant, or even a possibly ingestion of a proconvulsant.

The ideal consideration for flumazenil use is isolated iatrogenic BZD overdose in BZD-naive patients (eg, during conscious sedation on BZD-naive patient).

Questions & Answers

DDX

What are the differential diagnoses for Benzodiazepine Toxicity?

Medications

Which medications in the drug class Antidotes, Other are used in the treatment of Benzodiazepine Toxicity?

Contributor Information and Disclosures

Author

Chip Gresham, MD, FACEM Emergency Medicine Physician, Medical Toxicologist, and Intensive Care Consultant, Department of Emergency Medicine, Clinical Director of Medication Safety, Middlemore Hospital; Consultant Toxicologist, National Poisons Centre; Director, Auckland Regional Toxicology Service; Senior Lecturer, Auckland University Medical School, New Zealand

Chip Gresham, MD, FACEM is a member of the following medical societies: American College of Emergency Physicians, American College of Medical Toxicology, Australasian College for Emergency Medicine, Society for Academic Emergency Medicine

Disclosure: Nothing to disclose.

Chief Editor

Gil Z Shlamovitz, MD, FACEP Associate Professor of Clinical Emergency Medicine, Keck School of Medicine of the University of Southern California; Chief Medical Information Officer, Keck Medicine of USC

Gil Z Shlamovitz, MD, FACEP is a member of the following medical societies: American College of Emergency Physicians, American Medical Informatics Association

Disclosure: Nothing to disclose. https://emedicine.medscape.com/article/813255-print 7/8 12/10/2020 https://emedicine.medscape.com/article/813255-print Additional Contributors

Asim Tarabar, MD Assistant Professor, Director, Medical Toxicology, Department of Emergency Medicine, Yale University School of Medicine; Consulting Staff, Department of Emergency Medicine, Yale-New Haven Hospital

Disclosure: Nothing to disclose.

References

1. Soyka M. Treatment of Benzodiazepine Dependence. N Engl J Med. 2017 Mar 23. 376 (12):1147-1157. [Medline].

2. Ait-Daoud N, Hamby AS, Sharma S, Blevins D. A Review of Alprazolam Use, Misuse, and Withdrawal. J Addict Med. 2018 Jan/Feb. 12 (1):4-10. [Medline]. [Full Text].

3. Chyka PA, Seger D, Krenzelok EP, Vale JA. Position paper: Single-dose activated charcoal. Clin Toxicol (Phila). 2005. 43(2):61-87. [Medline].

4. Marraffa JM, Cohen V, Howland MA. Antidotes for toxicological emergencies: a practical review. Am J Health Syst Pharm. 2012 Feb 1. 69(3):199-212. [Medline].

5. Drug Abuse Warning Network, 2011: National Estimates of Drug-Related Emergency Department Visits. Substance Abuse and Mental Health Services Administration. Available at https://www.samhsa.gov/data/report/national-estimates-drug-related-emergency- department-visits-2004-2011-illicits-excluding. Accessed: February 7, 2019.

6. Substance Abuse and Mental Health Services Administration. SAMHSA’s Major Data Collections. SAMHSA. Available at https://www.samhsa.gov/samhsa-data-outcomes-quality/major-data-collections. May 16, 2018; Accessed: June 13, 2018.

7. Bachhuber MA, Hennessy S, Cunningham CO, Starrels JL. Increasing Benzodiazepine Prescriptions and Overdose Mortality in the United States, 1996-2013. Am J Public Health. 2016 Apr. 106 (4):686-8. [Medline]. [Full Text].

8. Gummin DD, Mowry JB, Spyker DA, Brooks DE, Beuhler MC, Rivers LJ, et al. 2018 Annual Report of the American Association of Poison Control Centers' National Poison Data System (NPDS): 36th Annual Report. Clin Toxicol (Phila). 2019 Dec. 57 (12):1220- 1413. [Medline]. [Full Text].

9. European Monitoring Centre of Drugs and Drug Addiction. European Monitoring Centre of Drugs and Drug Addiction. EMCDDA.europa.eu. Available at http://www.emcdda.europa.eu/publications/pods/benzodiazepines. June 2018; Accessed: February 7, 2019.

10. Shah NA, Abate MA, Smith MJ, Kaplan JA, Kraner JC, Clay DJ. Characteristics of alprazolam-related deaths compiled by a centralized state medical examiner. Am J Addict. 2012 Nov. 21 Suppl 1:S27-34. [Medline].

11. Isbister GK, O'Regan L, Sibbritt D, Whyte IM. Alprazolam is relatively more toxic than other benzodiazepines in overdose. Br J Clin Pharmacol. 2004 Jul. 58(1):88-95. [Medline]. [Full Text].

12. Jann M, Kennedy WK, Lopez G. Benzodiazepines: a major component in unintentional prescription drug overdoses with opioid analgesics. J Pharm Pract. 2014 Feb. 27(1):5-16. [Medline].

13. Darke S, Torok M, Duflou J. Circumstances and toxicology of sudden or unnatural deaths involving alprazolam. Drug Alcohol Depend. 2014 May 1. 138:61-6. [Medline].

14. [Guideline] Kleber HD, Weiss RD, Anton RF, et al. Treatment of patients with substance use disorders, second edition. American Psychiatric Association. Am J Psychiatry. 2006 Aug. 163(8 Suppl):5-82. [Medline]. [Full Text].

15. [Guideline] National Collaborating Centre for Mental Health. Self-harm: the short-term physical and psychological management and secondary prevention of self-harm in primary and secondary care. London (UK): National Institute for Clinical Excellence (NICE). 2004. 199. [Full Text].

16. Penninga EI, Graudal N, Ladekarl MB, Jürgens G. Adverse Events Associated with Flumazenil Treatment for the Management of Suspected Benzodiazepine Intoxication - A Systematic Review with Meta-Analyses of Randomised Trials. Basic Clin Pharmacol Toxicol. 2016 Jan. 118 (1):37-44. [Medline].

17. The Flumazenil in Benzodiazepine Intoxication Multicenter Study Group. Treatment of benzodiazepine overdose with flumazenil. Clin Ther. 1992 Nov-Dec. 14(6):978-95. [Medline].

18. Seger DL. Flumazenil--treatment or toxin. J Toxicol Clin Toxicol. 2004. 42(2):209-16. [Medline].

19. Lembke A, Papac J, Humphreys K. Our Other Prescription Drug Problem. N Engl J Med. 2018 Feb 22. 378 (8):693-695. [Medline]. [Full Text].

https://emedicine.medscape.com/article/813255-print 8/8 Journal of Affective Disorders 235 (2018) 198–205

Contents lists available at ScienceDirect

Journal of Affective Disorders

journal homepage: www.elsevier.com/locate/jad

Research paper A naturalistic examination of the perceived effects of cannabis on negative affect T ⁎ Carrie Cuttlera,b, , Alexander Spradlina, Ryan J. McLaughlina,b,c a Washington State University, Department of Psychology, P.O. Box 644820, Pullman, WA 99164-4820, USA b Translational Addiction Research Center, Washington State University, USA c Washington State University, Department of Integrative Physiology and Neuroscience, Pullman, WA 99164-7620, USA

ARTICLE INFO ABSTRACT

Keywords: Background: Cannabis is commonly used to alleviate symptoms of negative affect. However, a paucity of re- Cannabis search has examined the acute effects of cannabis on negative affect in everyday life. The current study provides Depression a naturalistic account of perceived changes in symptoms of depression, anxiety, and stress as a function of dose Anxiety and concentration of Δ9tetrahydrocannabinol (THC) and cannabidiol (CBD). Stress Method: Data from the app StrainprintTM (which provides medical cannabis users a means of tracking changes in Dose effects symptoms as a function of different doses and chemotypes of cannabis) were analyzed using multilevel mod- Multilevel modeling eling. In total, 11,953 tracked sessions were analyzed (3,151 for depression, 5,085 for anxiety, and 3,717 for stress). Results: Medical cannabis users perceived a 50% reduction in depression and a 58% reduction in anxiety and stress following cannabis use. Two puffs were sufficient to reduce ratings of depression and anxiety, while 10+ puffs produced the greatest perceived reductions in stress. High CBD (> 9.5%)/low THC (< 5.5%) cannabis was associated with the largest changes in depression ratings, while high CBD (> 11%)/high THC (> 26.5%) can- nabis produced the largest perceived changes in stress. No changes in the perceived efficacy of cannabis were detected across time. However, baseline symptoms of depression (but not anxiety or stress) appeared to be exacerbated across time/tracked sessions. Limitations: The primary limitations are the self-selected nature of the sample and the inability to control for expectancy effects. Conclusions: Cannabis reduces perceived symptoms of negative affect in the short-term, but continued use may exacerbate baseline symptoms of depression over time.

1. Introduction medical cannabis use currently recognize anxiety as a qualifying con- dition, and none overtly recognize depression or high levels of per- Cannabis is commonly used to alleviate depression, anxiety, and ceived stress as qualifying conditions for a medical cannabis card. This stress. Indeed, one of the most commonly reported motives for cannabis is largely because of a paucity of evidence for the efficacy of cannabis in use is to cope with stress (Hyman and Sinha, 2009), with 72% of daily treating negative affect. As such, the primary purpose of the present cannabis users reporting use of cannabis to relax or relieve tension study was to examine the perceived efficacy of cannabis in reducing (Johnston and O’ Malley, 1986). Further, recent research by symptoms of depression, anxiety, and stress in a naturalistic context. Sexton et al. (2016) revealed that the three most frequently endorsed While research on the acute effects of cannabis and its two primary reasons for medical cannabis use are for managing pain, anxiety, and constituents – Δ9tetrahydrocannabinol (THC) and cannabidiol (CBD) – depression, with over 58% of medical cannabis patients reporting use to on depression, anxiety, and stress is sparse, there is some limited evi- manage anxiety and over 50% reporting use for depression. Consistent dence from double-blind, placebo-controlled studies indicating that oral with these results, Webb and Webb (2014) found that 50% of medical CBD significantly reduced anxiety and discomfort during a simulated cannabis patients – who were using cannabis to treat pain – reported public speaking task in patients with social anxiety disorder that it provided relief from anxiety and stress. Nevertheless, only two of (Bergamaschi et al., 2011) and in healthy students (Zuardi et al., 1993). the 32 states, districts, and territories in the United States that permit Oral THC has similarly been shown to dose-dependently attenuate the

⁎ Corresponding author at: Washington State University, Department of Psychology, P.O. Box 644820, Johnson Tower, Pullman, WA 99164-4820, USA. E-mail address: [email protected] (C. Cuttler). https://doi.org/10.1016/j.jad.2018.04.054 Received 12 February 2018; Received in revised form 2 April 2018; Accepted 4 April 2018 Available online 06 April 2018 0165-0327/ © 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/BY-NC-ND/4.0/). C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205 subjective response to psychosocial stress (Childs et al., 2017). Further, emotionality (see Drevets et al., 2008 for review); therefore, regular use in patients being treated for fibromyalgia, regular use of nabilone (an of cannabis may have longer-term effects on negative affect (that may orally ingested synthetic cannabinoid receptor agonist) significantly be reversed following a period of abstinence). In the current study, we decreased anxiety but had no effects on depression (Skrabek et al., used archival data from StrainprintTM to specifically examine: 1) whe- 2008). Conversely, an oral dose of rimonabant (a cannabinoid receptor ther self-reported symptoms of depression, anxiety, and stress are sig- antagonist) has been shown to significantly increase depression nificantly reduced after using cannabis, 2) whether there are gender (Christensen et al., 2007) and anxiety (Bergamaschi et al., 2014; differences in these putative effects, 3) whether interactions between Christensen et al., 2007). THC and CBD predict symptom change, 4) whether symptom change It is important to note, however, that these effects have not always varies according to dose, 5) whether perceived efficacy of cannabis been consistent. Specifically, Pillard et al. (1974) found no significant changes over time, and 6) whether baseline (i.e., pre-cannabis use) effects of smoking low THC cannabis (1.4%) versus a placebo on an- symptoms of negative affect change over time. xiety following an anxiety-provoking film or public speaking task. Moreover, a handful of double-blind, placebo-controlled studies ex- 2. Method amining depression and anxiety as secondary outcomes in patients with other primary conditions (e.g., chronic pain, multiple sclerosis, cancer) 2.1. Procedure found no significant effects of nabilone (Frank et al., 2008), dronabinol (another orally ingested synthetic cannabinoid receptor agonist) To achieve our six aims, we obtained archival data from the medical (Narang et al., 2008), or nabiximol (a cannabis-based oromucosal cannabis app StrainprintTM. Using this free app, medical cannabis users spray) (Portenoy et al., 2012; Rog et al., 2005; Wade et al., 2004)on can track changes in the severity of their symptoms as a function of secondary symptoms of anxiety and depression. different chemotypes and doses of cannabis. Prior to using the app to Some of these conflicting results may pertain to the use of varying track their medical cannabis use, individuals enter basic demographic doses and differences in THC vs. CBD content. For instance, a recent information (i.e., gender and date of birth). Next, they enter all of their double-blind, placebo-controlled study with healthy adults revealed medical conditions and symptoms of those conditions by selecting from that low doses (7.5 mg) of oral THC attenuated the self-reported ne- a list of 279 conditions and 46 symptoms. They are further given the gative emotional effects of a psychosocial stressor, while high doses opportunity to enter information about the cannabis that they use by (12.5 mg) increased subjective distress, anxiety, and depression selecting from a list of products sold by licensed medical cannabis (Childs et al., 2017). Consistent with these results, Fusar- distributors in Canada. The THC and CBD content for each of these Poli et al. (2008) found that 10 mg of oral THC increased anxiety and products were obtained by analyses conducted by one of Health other negative emotions relative to a placebo, while oral CBD (600 mg) Canada's licensed dealers and is prepopulated within the app. It is im- led to a small decrease in anxiety that was not statistically significant in portant to note that Canada is somewhat unique to other countries in their small sample (n = 15, p = .06). that Health Canada enforces strict production guidelines, quality con- In the majority of studies described above, cannabinoids were ad- trol guidelines and mandatory lab testing from all ministry approved ministered orally. However, recent research indicates that only 8% of licensed dealers. This mandatory lab testing includes five stages of medical cannabis patients use oral administration (Sexton et al., 2016), processing; preparation, chromatography, general spectrometry, heavy while over 92% of medical and non-medical cannabis users report using metal spectrometry, and microbial analysis. Users also have the op- combustion/inhalation methods of administration (Schauer et al., portunity to enter additional product names and cannabinoid content 2016). Moreover, in many of these studies cannabinoids were ad- (% THC, % CBD) for products that are not prepopulated in the app. ministered prior to an objectively stressful task and participants were Users can subsequently track their medical cannabis sessions by: 1) asked to evaluate their affective state in response to that acute stressor. selecting the symptom(s) they are experiencing at the time, 2) rating As such, we have a limited understanding of the perceived efficacy of the severity of each symptom on a scale of 0 (none) to 10 (extreme), 3) cannabis in coping with feelings of negative affect in everyday life. selecting (or inputting) the product they will use, 4) indicating their Therefore, the primary objective of this study was to track the perceived method of administration (smoke, oil, vape, dab bubbler, dab portable, efficacy of inhaled cannabis in coping with feelings of negative affect in edible, pill, spray, transdermal, tincture), and 5) indicating the quantity medical cannabis users’ naturalistic environment. Furthermore, given of use (e.g., number of puffs ranging from 1 to 10 + ). Twenty minutes that symptoms of negative affect are more prevalent in women after use, individuals are prompted (via a push notification) to re-rate (American Psychiatric Association, 2013), and that women are more the severity of their symptom. likely to use cannabis to cope with symptoms of anxiety (Cuttler et al., For the present study, we obtained anonymous data from medical 2016), we further sought to examine potential gender differences in the cannabis users who used the app to treat symptoms of depression, an- perceived efficacy of cannabis in reducing symptoms of depression, xiety, and stress. Specifically, we obtained data on these individuals’ anxiety, and stress. anonymous ID codes; gender; ages; medical conditions and symptoms; The present study represents an attempt to complement the existing, self-reported symptom severity before and after each session of medical internally valid, double-blind, placebo-controlled studies, with a more cannabis use; duration of time between pre-and post-cannabis use ecologically valid, naturalistic approach. To this end, we obtained the symptom ratings; cannabinoid content (% THC, % CBD) for the can- global back-data from the app StrainprintTM, which offers medical nabis used in each session; as well as the method and quantity of use for cannabis users a means to track symptom severity before and after self- each session. The Office of Research Assurances determined that this medicating with cannabis. One advantage of this approach is that it anonymous archival study was exempt from the need for IRB review. allows us to examine the perceived effects of cannabis over time. In doing so, we can address two important questions. First, does the per- 2.2. Inclusion/Exclusion criteria ceived efficacy of cannabis in managing negative affect change over time? In other words, are the perceived effects of cannabis subject to We obtained data from 1,399 medical cannabis users who collec- tolerance? Second, does using cannabis to manage symptoms of de- tively used the app a total of 18,392 times to track changes in their pression, anxiety, and stress affect baseline symptoms of negative affect symptoms of depression, anxiety, or stress. Individuals’ use of the app to over time? Chronic cannabis use has been shown to cause down- track their medical cannabis sessions ranged from 1–972 sessions, with regulation of CB1 receptors in areas such as the prefrontal cortex, a mean of 13.15 (SD = 38.48) tracked sessions. Given potential dif- anterior cingulate cortex, hippocampus, and parahippocampal gyrus ferences in efficacy and onset across different routes of administration (Hirvonen et al., 2012), which are known to be implicated in mood and (e.g., oral vs. inhaled), only tracked sessions in which individuals

199 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205 indicated administering cannabis via one of five inhalation methods 3. Results (smoking, vaping, concentrates, dab bubbler, dab portable) were se- lected (n = 13,687; 74.42% of data). Tracked sessions that involved 3.1. Aim 1: overall change in symptom ratings administration via other methods (e.g., tincture, edibles) were ex- cluded. Given that the acute subjective effects of inhaled cannabis peak 3.1.1. Depression at about 10–30 minutes and taper off after 3–4h(Grotenhermen, 2003; Results of the first multilevel model revealed a significant reduction

Menkes et al., 1991), only the 11,953 tracked inhalation sessions for in ratings of depression from before (MBefore = 6.02, SE = 0.17) to after 2 those individuals who re-rated their symptoms within 4 h were in- (MAfter = 3.06, SE = 0.21) using cannabis, Wald χ (1, 560) = 364.08, cluded. The remaining inhalation sessions exceeded this time frame and p < .001. Further analyses revealed that depression symptom ratings were excluded. were reduced in 89.3% of tracked sessions, they were exacerbated in 3.2% of sessions, and there was no change in 7.5% of sessions.

2.3. Participants 3.1.2. Anxiety There was also a significant reduction in the ratings of anxiety fi 2 The nal sample comprised 11,953 tracked inhalation sessions. (MBefore = 5.98, SE = 0.12 vs. MAfter = 2.50, SE = 0.14), Wald χ (1, More specifically, 561 medical cannabis users (262 men, 299 women; 769) = 659.50, p < .001. Further, anxiety was reduced in 93.5% of age M = 33, SD = 10) used the app 3,151 times (Range 1–97; tracked sessions, they were exacerbated in 2.1% of sessions, and there M = 14.30, SD = 17.38) to track changes in depression. A total of 770 was no change in symptoms for 4.4% of sessions. users (363 men, 407 women; age M = 33, SD = 10) used the app 5,085 – times (Range 1 197; M = 23.38, SD = 35.62) to track changes in an- 3.1.3. Stress xiety. Finally, 726 people (323 men, 403 women; age M = 34, SD =9) Analysis of the stress model revealed a significant change in ratings used the app 3,717 times (Range 1–173; M = 18.93, SD = 28.37) to from before (MBefore = 5.99, SE = 0.13) to after (MAfter = 2.52, track changes in stress. SE = 0.14) using cannabis, Wald χ2 (1, 725) = 620.08, p < .001. Further, stress was reduced in 93.3% of tracked sessions, it increased in 2.7% of sessions, and there was no change in reported levels of stress for 2.4. Data analysis 4% of sessions.

Typically, in order to analyze data with both within- and between- 3.2. Aim 2: gender differences in change in symptom ratings subjects variability to consider, one would use a mixed-factorial ana- lysis of variance (ANOVA) or a multiple regression analysis. However, 3.2.1. Depression each of these techniques requires equal numbers of within-subjects As depicted in Fig. 1 (panel A), results indicated that both women, observations across participants. Given the differences in the number of Wald χ2 (1, 261) = 380.74, p < .001, and men, Wald χ2 = (1, sessions tracked using the app across individuals these approaches are 299) = 137.52, p < .001, reported a significant reduction in symptoms not appropriate. However, multilevel modeling (also known as linear of depression following cannabis use. There was no significant differ- mixed models, hierarchical linear models, or mixed-effects models) is a ence in the magnitude of change between the genders, Wald chi-square technique that permits the examination of change considering both (1, 261) = 0.02, p = .88. within- and between-subject variability despite differences in the number of observations across individuals. This technique involves estimating time-variant slope variables at the within-subject level that 3.2.2. Anxiety χ2 are then used to predict change at the between-subject level. The Both women, Wald (1, 362) = 582.32, p < .001 and men, Wald χ2 fi models tested in the present study were based on those presented in = (1, 407) = 244.61, p < .001, reported a signi cant reduction in Finch and Bolin (2016) with additional modifications and settings symptoms of anxiety following cannabis use. Comparisons of the gen- specified according to the guidelines provided by Muthén and ders indicated that women perceived a greater reduction in symptoms χ2 Muthén (2017). of anxiety than did men, Wald (1, 362) = 10.78, p < .001 (see Specifically, multilevel modeling was used to predict changes in Fig. 1, panel B). symptom severity as a function of gender, dose, and % THC/CBD. We also used multilevel modeling to examine changes in efficacy (i.e., 3.2.3. Stress 2 symptom reduction) and changes in baseline symptoms (i.e., pre-can- As shown in Fig. 1 (panel C) both women, Wald χ (1, 2 nabis use symptom ratings) across tracked sessions. All models were 322) = 274.45, p < .001, and men, Wald χ = (1, 403) = 412.58, tested with maximum likelihood estimation with robust standard errors p < .001, reported a significant reduction in stress after using can- using the latest version of Mplus (version 8; Muthén and nabis. There was no significant difference in the magnitude of symptom 2 Muthén, 2017). All predictor and outcome variables were modeled as change between the genders, Wald χ (1, 322) = 3.18, p = .07. functions of time/sessions at the within-subjects level, and the slopes of these regressions (i.e., regression coefficients) were used to test for the 3.3. Aim 3: THC × CBD effects on change in symptom ratings between-subjects level effects (e.g., for aim 3, time/sessions was used to predict % THC, % CBD, and symptom change at the within-subject le- 3.3.1. Depression vels, and the slope values produced at this level were used to test for the Models were also tested to examine whether the cannabinoid content effects of % THC and % CBD on symptom change at the between-sub- (% THC, % CBD) could predict change in reported symptom severity. The jects level). All estimates of slopes and intercepts were allowed to vary results of the model tested, to examine whether THC, CBD, and their in- randomly. For models that included interaction terms, the algorithm teraction predict change in symptoms of depression, revealed a significant used to estimate model parameters was set to numerical integration THC x CBD interaction, b = −0.03, p = .03. As depicted in Fig. 2 (panel A) with 7 integration points. the greatest reduction in ratings of depression were reported after using cannabis with relatively low levels of THC (one standard deviation [SD] below the mean of THC) and relatively high levels of CBD (one SD above the mean of CBD). See Table 1 for overall means and SDsforcannabisusedto treat depression, anxiety, and stress.

200 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205

Fig. 2. THC x CBD interactions predicting change in depression (panel A), Fig. 1. Symptom ratings of depression (panel A), anxiety (panel B), and stress anxiety (panel B), and stress (panel C). (panel C) before and after using cannabis in women and men with standard Note: Low = one SD below the mean value; High = one SD above the mean error bars. value. Overall means and SDs are provided in Table 1. Note: * denotes significant difference with p < .01. Table 1 3.3.2. Anxiety Overall means and standard deviations (SDs) of % THC and % CBD. In contrast, results of the model predicting change in symptoms of Depression Anxiety Stress anxiety, using THC, CBD and THC x CBD, revealed no significant in- teraction, b = −0.26, p = .55 (see Fig. 2, panel B). We then removed % THC Mean (SD) 15.76 (10.35) 15.29 (9.13) 16.53 (9.97) the interaction term from the model to test for main effects of THC and % CBD Mean (SD) 2.90 (6.73) 3.69 (7.93) 2.97 (8.36) CBD. Results revealed that neither THC, b = −0.05, p = .33, nor CBD content, b = −0.006, p = .80, were significant predictors of change in anxiety ratings. b = 1.47, p < .001. As shown in Fig. 2 (panel C), ratings of stress were reduced the most after using cannabis with relatively high levels of THC (one SD above the mean of THC) and relatively high levels of CBD (one SD 3.3.3. Stress above the mean of CBD). In contrast, there was no appreciable differences in ff The multilevel model testing the e ects of cannabinoid content on symptom change following use of cannabis with high THC/low CBD, low changes in ratings of stress revealed a significant THC x CBD interaction,

201 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205

Fig. 3. Change in depression (panel A), anxiety (panel B), and stress (panel C) across different doses (1 to 10 + puffs) of cannabis with standard error bars.

Table 2 Multilevel models testing curvilinear relationships between dose and change in ratings of symptoms of anxiety.

Model 1 (Dose2) Model 2 (Dose3) Model 3 (Dose4)

Predictors bSEpPredictors bSEpPredictors bSEp

Dose 0.17 0.15 .26 Dose 0.17 0.14 .24 Dose 0.17 0.15 .25 Dose2 0.74 0.33 .02 Dose2 0.31 0.14 .04 Dose2 0.21 0.11 .04 Dose3 0.40 0.18 .03 Dose3 0.23 0.11 .04 Dose4 0.29 0.13 .03 Intercept −0.005 0.01 .34 Intercept −0.005 0.01 .34 Intercept −0.005 0.01 .33

Note: b = unstandardized regression coefficient, SE = standard error of estimate.

202 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205

THC/high CBD, or low THC/low CBD. than men to report using cannabis to manage anxiety (Cuttler et al., 2016). Across both genders, self-reported symptoms of depression were 3.4. Aim 4: effects of dose on change in symptom ratings reduced by approximately 50%, while symptoms of anxiety and stress were reduced by approximately 58%. Moreover, for the vast majority of 3.4.1. Depression tracked sessions, users reported a reduction in symptoms of depression Several multilevel models were tested to examine the impact of dose (89%), anxiety (93%), and stress (93%) after inhaling cannabis. It is on change in depression symptom ratings. Results revealed a non- important to note that these percentages are likely inflated in the pre- significant linear effect of dose predicting change, b = 0.06, p = .75. sent sample, as individuals who regularly experience symptom ex- We therefore tested for curvilinear relationships (i.e., dose2, dose3, acerbation following use of cannabis may be less likely to continue to dose4); however, none of these models revealed significant effects of use cannabis to treat their symptoms and/or to track their symptom dose on change in symptoms of depression (see Fig. 3, panel A). changes across time. Nevertheless, results from the present study are consistent with the reported anxiolytic, stress-alleviating effects of 3.4.2. Anxiety cannabis, and suggest that users experience significant and substantial Results of models testing change in ratings of anxiety across dif- reductions in symptoms of negative affect following cannabis use. ferent doses also revealed a nonsignificant linear effect, b = 0.15, Results from the multilevel models tested to examine THC x CBD p = .33. We therefore tested several models to explore curvilinear re- interactions revealed a significant cross-over interaction predicting lationships. First, a model was tested wherein we added the quadratic change in ratings of depression. Low levels of THC combined with high term for dose (i.e., dose2), and we found a significant curvilinear re- levels of CBD predicted the greatest reductions in reported symptoms of lationship (see Table 2, Model 1). Another model was tested wherein depression, while high levels of THC combined with high levels of CBD we added a cubic term for dose (i.e., dose3) to the previously tested predicted the lowest levels of reported symptom reduction. In contrast, model, and parameter estimates remained significant (see Table 2, varying levels of THC (high vs. low) appeared to have little influence on Model 2). Lastly, a model was tested wherein we added dose to the the degree of symptom reduction when CBD was low. Similarly, varying fourth power (i.e., dose4). Once again, results revealed a significant, levels of THC (high vs. low) appeared to have little influence on change curvilinear relationship (see Table 2, Model 3). Further contrasts re- in stress ratings when CBD was low. However, when CBD was high, vealed that 1 puff produced significantly smaller changes in ratings of there was an effect of THC such that higher levels of THC predicted anxiety than all other doses (2 to 10+), but no other differences across greater reductions in symptoms of stress relative to low levels of THC. doses beyond 1 puff were detected (see Fig. 3, panel B). These results suggest that cannabis with relatively high CBD (e.g., > 9.5%) and low THC (e.g., < 5.5%) is perceived to be more effective in 3.4.3. Stress reducing symptoms of depression, while cannabis with high CBD The model tested to examine the effect of varying doses on change (e.g., > 11%) and high THC (e.g., > 26.5%) is perceived to be more in ratings of stress revealed a significant linear effect of dose, b = 0.45, effective in reducing stress. The non-medical cannabis market is cur- p = .03. As depicted in Fig. 3 (panel C), further contrasts revealed the rently dominated by the sales of high THC cannabis products following significant differences in doses: 1 puff <5,6,7,8,and (Smart et al., 2017) but these results suggest that CBD is an important 10 + puffs; 2 puffs < 5, 6, 7, 8, and 10 puffs; 10 puffs > 9, 6, 5, 4, 3, component of cannabis and that medical cannabis users should seek out 2, and 1 puffs. cannabis with CBD levels of 10% or higher. While intriguing, there are currently no controlled examinations of THC x CBD interactions in the 3.5. Aim 5: changes in perceived efficacy of cannabis across tracked treatment of depression or stress, and it is possible that anecdotal evi- sessions dence propagated by bud-tenders and popular culture could be biasing medical cannabis users’ expectations and experiences. That is, medical Results of the multilevel analyses examining changes in perceived cannabis users’ beliefs regarding the therapeutic efficacy of THC- and/ efficacy of cannabis (i.e., tolerance effects) across time/sessions re- or CBD-rich chemotypes could have contributed to a potential ex- vealed no significant change in the perceived efficacy of cannabis on pectancy effect. Future studies should examine this possibility in a more depression (b = 0.003, p = .52), anxiety (b = 0.007, p = .07), or stress controlled setting. (b = 0.007, p = .32) across tracked sessions. The results also indicate the presence of a positive, linear, re- lationship between dose and perceived changes in stress. 10+ puffs 3.6. Aim 6: changes in baseline symptom ratings across tracked sessions were associated with the greatest change in ratings of stress. In contrast, models tested to examine the effects of dose on perceived changes in Results of the multilevel analysis examining change in baseline symptoms of anxiety revealed the presence of significant curvilinear symptom ratings (i.e., ratings of depression immediately before using relationships. Follow-up tests revealed that 1 puff was perceived as less cannabis) across tracked sessions indicated that baseline symptoms of effective than any other dose; however, there were no other significant depression significantly increased across time/sessions, b = 0.008, differences across any other doses (e.g., 2 puffs were perceived to be as p = .006. In contrast, the analyses examining change in baseline effective as 10+ puffs). Finally, we found no evidence for dose effects symptom ratings of anxiety and stress across tracked sessions indicated on change in ratings of depression. In other words, a single puff resulted no significant changes in baseline symptoms of anxiety (b = 0.007, in the same magnitude of change in ratings of depression as 10+ puffs. p = .09), or stress, (b = 0.01, p = .26). These findings may support the notion of “micro-dosing” to alleviate symptoms of depression and anxiety. Nevertheless, it is important to 4. Discussion note that different methods of administration (e.g., smoking, con- centrate, dab bubbler) were included together in these analyses and The present study was conducted to provide a naturalistic account these different methods of administration would affect the potency of of perceived changes in symptoms of negative affect as a function of the cannabis. Therefore, future research is needed to manipulate cannabis use. The results indicate that both women and men perceived method of administration, and then contrast the relative effects of dif- a significant reduction in symptoms of depression, anxiety, and stress ferent doses of cannabis, to provide more precise guidance on optimal after inhaling cannabis. Despite comparable levels of anxiety before doses for different routes of administration. using cannabis, women reported a significantly greater decrease in Collectively, these results appear to suggest that in the short-term, anxiety following cannabis consumption compared to men. This is cannabis effectively reduces perceived levels of depression, anxiety, somewhat consistent with previous findings that women are more likely and stress in both women and men. But what, if any, longer-term

203 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205 consequences are associated with the repeated use of cannabis to Nevertheless, these limitations are offset by several noteworthy manage states of negative affect? A major advantage of the multilevel strengths. Namely, since the data were obtained from a large sample of modeling approach is that it affords exploration of change in perceived medical cannabis users who were using a variety of cannabis products efficacy of cannabis in individuals over time. The results of these ana- in their natural environment, the study has very high ecological va- lyses revealed no apparent subjective tolerance effects, which is con- lidity, and the results are likely to reflect the actual experiences of sistent with recent evidence of no objective tolerance to the psycho- people who use cannabis to treat symptoms of negative affect. motor effects of THC (Ramaekers et al., 2016). Moreover, medical cannabis users’ motivation for using the app is Finally, examination of whether repeated use of cannabis to manage predominantly to track their personal symptoms to better understand states of negative affect results in any appreciable change in baseline the products and doses of cannabis that produce the most beneficial (pre-cannabis use) symptoms over time indicated that baseline ratings effects for them. Although Strainprint's™ terms of use indicate that the of anxiety and stress remained fairly stable across tracked sessions, data may be used for any purpose deemed appropriate, most users while baseline ratings of depression significantly increased over time/ would be unaware that their data are being used for scientific in- sessions. The value of the regression coefficient indicates that for every vestigations. Therefore, it is unlikely that the results are biased by users’ additional tracked session over time, one would predict a 0.008-unit explicit motivations to portray cannabis in a good light. increase in baseline ratings of depression (i.e., after 125 treatment In summary, the internal validity of the findings of the present study sessions, one would predict a 1-unit increase in baseline depression may be threatened by implicit biases (i.e., expectancy effects), but the ratings on a 0 to 10 scale). This is consistent with recent evidence in- findings are unlikely to be threatened by explicit biases and have high dicating that using cannabis to cope with distress is associated with ecological validity. As such, results from the present study provide an more cannabis-related problems and increased symptoms of depression important complement to the more internally valid, controlled labora- (Bonn-Miller et al., 2014; Moitra et al., 2015). Chronic cannabis use tory studies. decreases CB1 receptor availability in cortical areas implicated in mood disorders (Hirvonen et al., 2012), and a growing body of preclinical 4.2. Conclusions evidence indicates that genetic or pharmacological CB1 receptor blockade produces a phenotype that is strikingly reminiscent of the Results from the present study indicate that medical cannabis users fi symptom pro le of major depression (see Gorzalka and Hill, 2011 for report a substantial and significant reduction in symptoms of negative review). Collectively these results suggest that chronic use of cannabis affect shortly after using cannabis. Importantly, while acute cannabis to cope with symptoms of depression may increase susceptibility for intoxication temporarily alleviates perceived states of depression, an- depression by altering the endocannabinoid system. Fortunately, al- xiety, and stress, the repeated use of cannabis does not appear to lead to terations in CB1 receptor availability in chronic cannabis users are re- any longer-term reductions in these symptoms. versible after only a short (∼2 day) period of abstinence, with no sig- nificant differences after 28 days of abstinence (D’ Souza et al., 2016). Contributors Finally, it is worthwhile to note that there is evidence that anti- depressant medications are effective in the short-term, but that longer Carrie Cuttler conceived of the idea and research questions, ob- duration of use may actually increase vulnerability to relapse upon tained the data, assisted with analyses, and prepared the first complete discontinuation (Fava, 2003). Thus, similar to more conventional draft of the manuscript. Alexander Spradlin helped to conceive the re- pharmacological treatments, cannabis may temporarily mask symptoms search questions, conducted the analyses, and assisted in writing the of negative affect but may not effectively reduce these symptoms in the results section. Ryan J. McLaughlin helped to conceive the research long-term. questions, created the figures, aided in the interpretation of the results, and contributed to the writing of the manuscript. All authors approved 4.1. Limitations and strengths the final article. One limitation of the present study is that the sample likely un- derrepresents individuals who do not find cannabis to be an effective Funding means for reducing their symptoms of negative affect; such individuals would be unlikely to continue to use cannabis for this purpose. Another This work was supported by Washington State University's Alcohol limitation is the lack of a placebo control group. Given that the data and Drug Abuse Research Program (Dedicated Marijuana Account). The were obtained from medical cannabis users who were using their own funder had no role in the conduct or results of the study. cannabis in their own environment, it was not possible to obtain a comparison group. As such, it is possible – and likely – that at least Declarations of interest some of the detected effects are driven by expectations individuals have about the efficacy of cannabis for treating states of negative affect. None. Therefore, it is vital that the results from the present study be further investigated under double-blind, placebo-controlled conditions. Acknowledgments While the majority of the data on THC and CBD levels were obtained from licensed producers who are held to strict testing standards by Health We would like to thank the creators of StrainprintTM for freely and Canada, some of these data were entered by users of the app. As such some openly providing the data used in this study. of these data may have more questionable reliability. Future more con- trolled research involving the testing of a select few products is therefore fl needed to further probe potential THC x CBD interactions. Finally, the Con ict of interest failure to consider content of other phytocannabinoids and terpenoids found in cannabis represents a limitation of the present study. There are over 100 None. additional phytocannabinoids and at least 120 terpenes in cannabis that may contribute to its medicinal properties, either independently, or by ex- Supplementary materials acerbating/mitigating the pharmacological effects of THC (Calvi et al., 2018). The effects of these other compounds will need to be explored in Supplementary material associated with this article can be found, in future research when these data become more readily available. the online version, at doi:10.1016/j.jad.2018.04.054.

204 C. Cuttler et al. Journal of Affective Disorders 235 (2018) 198–205

References Pharmacokinet. 42, 327–360. http://dx.doi.org/10.2165/00003088-200342040- 00003. Hirvonen, J., Goodwin, R.S., Li, C.T., Terry, G.E., Zoghbi, S.S., Morse, C., Pike, V.W., American Psychiatric Association., 2013. Diagnostic and Statistical Manual of Mental Volkow, N.D., Huestis, M.A., Innis, R.B., 2012. Reversible and regionally selective fi Disorders, fth ed. American Psychiatric Publishing, Arlington, VA. downregulation of brain cannabinoid CB1 receptors in chronic daily cannabis smo- Bergamaschi, M.M., Queiroz, R.H.C., Chagas, M.H.N., de Oliveira, D.C.G., De Martinis, kers. Mol. Psychiatry 17, 642–649. http://dx.doi.org/10.1038/mp.2011.82. B.S., Kapczinski, F., Quevedo, J., Roesler, R., Schröder, N., Nardi, A.E., Martín-Santos, Hyman, S.M., Sinha, R., 2009. Stress-related factors in cannabis use and misuse: im- R., Hallak, J.E., Zuardi, A.W., Crippa, J.A.S., 2011. Cannabidiol reduces the anxiety plications for prevention and treatment. J. Subst. Abuse Treat. 36, 400–413. http:// induced by simulated public speaking in treatment-naïve social phobia patients. dx.doi.org/10.1016/j.jsat.2008.08.005. – Neuropsychopharmacology 36, 1219 1226. http://dx.doi.org/10.1038/npp.2011.6. Johnston, L.D., O'Malley, P.M., 1986. Why do the nations’ students use drugs and alcohol? Bergamaschi, M.M., Queiroz, R.H.C., Chagas, M.H.N., Linares, I.M.P., Arrais, K.C., de Self-reported reasons from nine national surveys. J. Drug Issues 16, 29–66. http://dx. Oliveira, D.C.G., Queiroz, M.E., Nardi, A.E., Huestis, M.A., Hallak, J.E.C., Zuardi, doi.org/10.1177/002204268601600103. ff A.W., Moreira, F.A., Crippa, J.A.S., 2014. Rimonabant e ects on anxiety induced by Menkes, D.B., Howard, R.C., Spears, G.F., Cairns, E.R., 1991. Salivary THC following simulated public speaking in healthy humans: a preliminary report. Hum. cannabis smoking correlates with subjective intoxication and heart rate. – Psychopharmacol. Clin. Exp. 29, 94 99. http://dx.doi.org/10.1002/hup.2374. Psychopharmacology 103, 277–279. Bonn-Miller, M.O., Boden, M.T., Bucossi, M.M., Babson, K.A., 2014. Self-reported can- Moitra, E., Christopher, P.P., Anderson, B.J., Stein, M.D., 2015. Coping-motivated mar- nabis use characteristics, patterns and helpfulness among medical cannabis users. ijuana use correlates with DSM-5 cannabis use disorder and psychological distress – Am. J. Drug Alcohol Abuse 40, 23 30. http://dx.doi.org/10.3109/00952990.2013. among emerging adults. Psychol. Addict Behav. 29, 627–632. http://dx.doi.org/10. 821477. 1037/adb0000083. Calvi, L., Pentimalli, D., Panseri, S., Giupponi, L., Gelmini, F., Beretta, G., Vitali, D., Muthén, L.K., Muthén, B.O., 2017. Mplus User's Guide, eighth ed. Muthén & Muthén, Los Bruno, M., Zilio, E., Pavlovic, R., Giorgi, A., 2018. Comprehensive quality evaluation Angeles, CA. fl of medical Cannabis sativa L. in orescence and macerated oils based on HS-SPME Narang, S., Gibson, D., Wasan, A.D., Ross, E.L., Michna, E., Nedelijkovic, S.S., Jamison, ® coupled to GC-M and LC-HRMS (q-exactive orbitrap ) approach. J. Pharm. Biomed. R.N., 2008. Efficacy of dronabinol as an adjuvant treatment for chronic pain patients – Anal. 150, 208 219. http://dx.doi.org/10.1016/j.jpba.2017.11.073. on opioid therapy. J. Pain 9. , 254-254. doi:10.1016/j.jpain.2007.10.018. ff Childs., E., Lutz, A.A., de Wit, H., 2017. Dose-related e ects of delta-9-THC on emotional Pillard, R.C., McNair, D., Fisher, S., 1974. Does marijuana enhance experimentally in- responses to acute psychosocial stress. Drug Alcohol Depend. 177. , 136.144. doi:10. duced anxiety? Psychopharmacologia 40, 205–210. http://dx.doi.org/10.1007/ 1016/j.drugalcdep.2017.03.030. BF00429414. ffi Christensen, R., Kristensen, P.K., Bartels, E.M., Bliddal, H., Astrup, A., 2007. E cacy and Portenoy, R.K., Ganae-Motan, E.D., Allende, S., Yanagihara, R., Shaiova, L., Weinstein, S., safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. McQuade, R., Wright, S., Fallon, M.T., 2012. Nabiximols for opioid-treated cancer – Lancet 370, 1706 1713. http://dx.doi.org/10.1016/S0140-6736(07)61721-8. patients with poorly-controlled chronic pain: a randomized placebo-controlled, ff ff Cuttler, C., Mischley, L., Sexton, M., 2016. Sex di erences in cannabis use and e ects: a graded-dose trial. J. Pain 13, 438–449. http://dx.doi.org/10.1016/j.pain.2012.01. – cross-sectional survey of cannabis users. Cannabis Cannabinoid Res. 1, 166 175. 003. http://dx.doi.org/10.1089/can.2016.0010. Ramaekers, J.G., van Wel, J.H., Spronk, D.B., Toennes, S.W., Kuypers, K.P.C., Theunissen, Drevets, W.C., Price, J.L., Furey, M.L., 2008. Brain structural and functional abnormalities E.L., Verkes, R.J., 2016. Cannabis and tolerance: acute drug impairment as a function in mood disorders: implications for neurocircuitry models of depression. Brain Struct. of cannabis use history. Sci. Rep. 6, 26843. http://dx.doi.org/10.1038/srep26843. – Funct. 213, 93 118. http://dx.doi.org/10.1007/s00429-008-0189-x. Rog, D.J., Nurmikko, T.J., Friede, T., Young, C.A., 2005. Randomized controlled trial of D'Souza, D.C., Cortes-Briones, J.A., Ranganathan, M., Thurnauer, H., Creatura, G., Surti, cannabis-based medicine in central pain in multiple sclerosis. Neurology 65, T., Planeta, B., Neumeister, A., Pittman, B., Normandin, M., Kapinos, M., Ropchan, J., 812–819. http://dx.doi.org/10.1212/01.wnl.0000176753.45410.8b. Huang, Y., Carson, R.E., Skosnik, P.D., 2016. Rapid changes in CB1 receptor avail- Schauer, G.L., King, B.A, Bunnell, R.E., Promoff, G., McAfee, T.A., 2016. Toking, vaping, ability in cannabis-dependent males after abstinence from cannabis. Biol. Psychiatry and eating for health or fun: marijuana use patterns in adults, U.S. Am J Prev Med 50, – Cogn. Neurosci. Neuroimaging 1, 60 67. http://dx.doi.org/10.1016/j.bpsc.2015.09. 1–8. http://dx.doi.org/10.1016/j.amepre.2015.05.027. 008. Sexton, M., Cuttler, C., Finnell, J., Mischley, L., 2016. A cross-sectional survey of medical Fava, G.A., 2003. Can long-term treatment with antidepressant drugs worsen the course cannabis users: patterns of use and perceived efficacy. Cannabis Cannabinoid Res. 1, – of depression. J. Clin. Psychiatry 64, 123 133. http://dx.doi.org/10.4088/JCP. 131–138. http://dx.doi.org/10.1089/can.2016.0007. v64n0204. Skrabek, R.Q., Galimova, L., Ethans, K., Perry, D, 2008. Nabilone for the treatment of pain fi Finch, W.H., Bolin, J.E., 2016. Multilevel Modeling Using Mplus, rst ed. Chapman Hall/ in fibromyalgia. J. Pain 9, 164–173. http://dx.doi.org/10.1016/j.jpain.2007.09.002. CRC Statistics in the Social and Behavioral Sciences, Boca Raton, FL. Smart, R., Caulkins, J.P., Kilmer, B., Davenport, S., Midgette, G., 2017. Variation in Frank, B., Serpell, M.G., Hughes, J., Matthews, J.N.S., Kapur, D., 2008. Comparisons of cannabis potency & prices in newly-legal market: evidence from 30 million cannabis ff analgesic e ects and patient tolerability of nabilone and dihydrocodeine for chronic sales in Washington State. Addiction 112, 2167–2177. http://dx.doi.org/10.1111/ – neuropathic pain: randomized, crossover, double blind study. BMJ 336, 199 201. add.13886. http://dx.doi.org/10.1136/bmj.39429.619653.80. Wade, D.T., Makela, P., Robson, R., House, H., Bateman, C., 2004. Do cannabis-based Fusar-Poli, P., Crippa, J.A., Bhattacharyya, S., Borgwardt, S.J., Allen, P., Martín-Santos, medicinal extracts have general or specificeffects on symptoms in multiple sclerosis? R., Seal, M., Surguladze, S.A., O'Carrol, C., Atakan, Z., Zuardi, A.W., McGuire, P.K., A double-blind, randomized, placebo-controlled study on 160 patients. Mult. Scler. ff Δ9 2008. Distinct e ects of -tetrahydrocannabinol and cannabidiol on neural activa- 10, 434–441. http://dx.doi.org/10.1191/1352458504ms1082oa. – tion during emotional processing. Arch. Gen. Psychiatry 66, 95 105. http://dx.doi. Webb, C.W., Webb, S.M., 2014. Therapeutic benefits of cannabis: a patient survey. Hawaii org/10.1001/archgenpsychiatry.2008.519. J. Med. Public Health 73, 109–111. Gorzalka, B.B., Hill, M.N., 2011. Putative role of endocannabinoid signaling in the Zuardi, A.W., Cosme, R.A., Graeff, F.G., Guimarães, F.S., 1993. Effects of ipsapirone and etiology of depression and actions of antidepressants. Prog. Neuropsychopharmacol. cannabidiol on human experimental anxiety. J. Psychopharmacol. 7, 82–88. http:// – Biol. Psychiatry 35, 1575 1585 doi: 10/1016/j.pnpbp.2010.11.021. dx.doi.org/10.1177/026988119300700112. Grotenhermen, F., 2003. Pharmacokinetics and pharmacodynamics of cannabinoids. Clin.

205 NIH Public Access Author Manuscript Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25.

NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Recent Pat CNS Drug Discov. 2012 April 1; 7(1): 25–40.

Cannabinoid-related agents in the treatment of anxiety disorders: current knowledge and future perspectives

Simone Tambaro and Marco Bortolato Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles (CA), USA

Abstract Rich evidence has shown that cannabis products exert a broad gamut of effects on emotional regulation. The main psychoactive ingredient of hemp, Δ9-tetrahydrocannabinol (THC), and its synthetic cannabinoid analogs have been reported to either attenuate or exacerbate anxiety and fear-related behaviors in humans and experimental animals. The heterogeneity of cannabis- induced psychological outcomes reflects a complex network of molecular interactions between the key neurobiological substrates of anxiety and fear and the endogenous cannabinoid system, mainly consisting of the arachidonic acid derivatives anandamide and 2-arachidonoylglycerol (2-AG) and two receptors, respectively termed CB1 and CB2. The high degree of interindividual variability in the responses to cannabis is contributed by a wide spectrum of factors, including genetic and environmental determinants, as well as differences in the relative concentrations of THC and other alkaloids (such as cannabidiol) within the plant itself. The present article reviews the currently available knowledge on the herbal, synthetic and endogenous cannabinoids with respect to the modulation of anxiety responses, and highlights the challenges that should be overcome to harness the therapeutic potential of some of these compounds, all the while limiting the side effects associated with cannabis consumption.

Keywords cannabis; anxiety; CB receptors; endocannabinoids; Δ9-tetrahydrocannabinol; cannabidiol

INTRODUCTION Anxiety is generally defined as an emotional state characterized by maladaptive and excessive emotional responsiveness to potentially dangerous circumstances. The pathological expression of anxiety leads to enduring emotional perturbations with a consistent apprehension towards the possibility of future, vaguely defined negative events [1]. According to the current classification of anxiety disorders in the fourth edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) [2], the main diagnostic entities in this category are: - generalized anxiety disorder (GAD), featuring general irritability, anxiety attacks, chronic apprehension/anxious expectation and secondary phobic avoidance.

Corresponding author: Marco Bortolato, MD PhD, Dept. of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Ave PSC 527, Los Angeles, CA 90089, Phone: 323-442-3225, Fax: 323-442-3229, [email protected]. Tambaro and Bortolato Page 2

- panic disorder, characterized by brief (2-10 min) spells of overwhelming anxiety or fear, accompanied by somatic and cognitive symptoms;

NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript - social anxiety disorder (or social phobia), defined as extreme agitation in social contexts and avoidance of social situations; - obsessive-compulsive disorder (OCD), characterized by recurrent and intrusive anxiogenic thoughts (obsessions), and stereotyped behaviors (compulsions) aimed at the reduction of the distress caused by the obsessions. - post-traumatic stress disorder (PTSD), in which a prior intense trauma results in a long-lasting anxious response, with re-experiencing/flashback phenomena, avoidance and emotional numbing. In keeping with their different clinical features and phenomenological presentations, these disorders are underpinned by divergent neurobiological alterations and respond to partially different pharmacotherapeutic strategies (outlined in Table 1). A fundamental contribution in our understanding of the neural bases of anxiety disorders and in the development of novel therapies has been afforded by animal models and testing paradigms for anxiety-like behaviors (summarized in Table 2).

Over the last decades, converging epidemiological, clinical and preclinical data have pointed to a key implication of cannabis and its endogenous system in the regulation of anxiety. In the following sections, we will present a brief synopsis on cannabinoids and the available classes of related agents, with a specific focus on their anxiolytic potential, and the scientific challenges that should be overcome to fully establish the applicability of such drugs in the therapy of anxiety disorders.

HERBAL AND SYNTHETIC CANNABINOIDS Herbal cannabinoids The three species included in the Cannabis genus (or sub-species, depending on the taxonomic classification; see [3], for a detailed discussion on the issue), sativa, indica and ruderalis, feature at least 85 unique terpenophenolic compounds, collectively named phytocannabinoids [4]. The main classes of phytocannabinoids are outlined in Figure 1. Quantitative analyses of cannabis constituents are usually performed by chromatographic techniques (generally Gas Chromatography, but also Thin-Layer Chromatography, or High- Performance Liquid Chromatography), often coupled with Mass Spectrometry. A detailed description of the instrumental methods used for classification and source tracing of Cannabis products (including DNA identification for forensic and intelligence purposes) is beyond the scope of this review, but can be found in [5-7].

The chemical fingerprinting of hemp products has revealed that the two most abundant phytocannabinoids are Δ9-tetrahydrocannabinol (THC, also named dronabinol) and cannabidiol (CBD):

The main psychoactive constituent of Cannabis, THC is a highly lipophilic alkaloid produced mainly in the leaves, flowers and glandular trichomes of the plant. Most of the pharmacological effects elicited by hemp products, including emotional and cognitive changes, analgesia, hypothermia and appetite stimulation, are considered to be reflective of the action of THC as a partial agonist of cannabinoid CB1 and CB2 receptors (see below). Additionally, THC has been shown to act as an acetylcholinesterase inhibitor [8-10].

In contrast with THC, CBD is not psychotropic, but has nevertheless been shown to play a role in the modulation of behavioral effects of cannabis [11]. In fact, the THC: CBD ratio is

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 3

the main criterion to define different cannabis chemotypes [12] and has been posited to contribute to the variability in neurobehavioral outcomes of marijuana or hashish

NIH-PA Author Manuscript NIH-PA Author Manuscriptconsumption NIH-PA Author Manuscript [13,14]. Interestingly, most cannabis strains encountered in the illegal markets generally have elevated amounts of THC [15].

The different characteristics of THC and CBD are underpinned by their distinct mechanisms of action. Whereas THC has nanomolar affinity for both CB1 (Ki = 25.1 nmol/L) and CB2 (Ki = 35.2 nmol/L) receptors, CBD exhibits much lower affinity for either target [16-20]; however, the latter phytocannabinoid was recently found to act as a highly potent antagonist/ inverse agonist of both CB receptors [21], possibly due to a non-competitive mechanism of receptor blockade [22]. Additionally, CBD has been shown to exert some of its actions through other receptors, including the vanilloid receptor VR1 and the serotonin receptor 5- HT1A (for a general overview of the topic, see [11]).

The other main phytocannabinoids, including cannabigerol (CBG), cannabichromene (CBC) and cannabinol (CBN) (Fig. 1) [4,23], have been shown to exert antibiotic and antiinflammatory properties, but have not been strongly associated with the behavioral effects of Cannabis; nevertheless, the recent discovery that CBG is a highly potent agonist for α2 adrenoceptor and a blocker of serotonin 5-HT1A receptor [24] underscores the potential importance of these and other alkaloids in the psychoactive profile of cannabis.

Synthetic cannabinoids In addition to phytocannabinoids, several classes of synthetic CB receptor agonists have been developed; among these families, the best characterized are the synthetic analogs of THC - such as the biciclic compounds CP 47,497, CP 55,244, CP 55,940 and the benxopyrans HU-210 and nabilone (Fig. 2) - and the aminoalkylindole derivatives - including WIN 55,212-2, JWH-015, JWH-018, JWH-073, JWH-081 and JWH-398 (for a general review, see [23]). Of these agents, only nabilone has been approved for clinical use as an antiemetic treatment and an adjunct analgesic for neuropathic pain [25]. Other more potent synthetic cannabinoids, such as CP 47,497, HU-210 and most JWH compounds, have regrettably gained great popularity in the market of recreational substances during the last decade, under the generic brand names of “Spice” or “K2”. Unlike THC, which is a partial agonist of CB1 receptors, these agents are full, high-potency CB1 receptor activators [26,27], thereby eliciting greater psychotropic effects than THC (as CB1 receptors are the key mediators of the psychotropic actions of cannabis). This characteristic, together with their legal status (recently revoked across most Western countries, including USA as of March 2011) and lack of available testing procedures for the detection of urinary metabolites, has unfortunately contributed to the great diffusion of “Spice” blends in Central and Western Europe, as well as Australasia.

ENDOCANNABINOIDS AND THEIR RECEPTORS Following the identification of THC in the 1960s [28], extensive research was devoted to the identification of its biological targets and endogenous counterparts. Both objectives were met around 30 years later, with the characterization of the two major cannabinoid receptors, CB1 [29] and CB2 [30] as well as the discovery of two most prominent endocannabinoids N- arachidonoylethanolamine (commonly named anandamide from the Sanskrit ānanda, bliss) [31] and 2-arachidonoylglycerol (2-AG) [32,33] (Fig. 3).

CB receptors

Although CB1 and CB2 receptors only share 44% sequence identity (68% in the transmembrane domains), they are both coupled to Gi/o proteins [34] and activated by both anandamide and 2-AG. In line with their metabotropic nature, CB receptors mediate their

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 4

intracellular response through a number of changes affecting signaling cascades, such as inhibition of adenylyl cyclase, activation of G-protein-activated inwardly rectifying

NIH-PA Author Manuscript NIH-PA Author Manuscriptpotassium NIH-PA Author Manuscript channels (GIRKs) and phosphorylation of extracellular signal-related kinases (ERKs) [35,36]. The distribution pattern of CB1 and CB2 receptors is strikingly divergent, indicating diverse physiological functions: CB1 is the most abundant metabotropic receptor in the brain, and is primarily distributed in the synaptic terminals of neurons across all the major structures that regulate emotional responsiveness, perception and memory, including prefrontal cortex, amygdala, septo-hippocampal system, striatum, thalamus, brainstem nuclei etc. [37-41]. CB1 receptors are typically located on presynaptic terminals [42,43], but they have also been identified in postsynaptic locations [44,45]. Presynaptic CB1 receptors are posited to serve critical functions for the regulation of synaptic plasticity and neurotransmitter release; in particular, they mediate the depolarization-induced suppression of inhibition (DSI) and depolarization-induced suppression of excitation (DSE), consisting in the reduction of γ-amino-butyric acid (GABA) or glutamate release, respectively, from presynaptic boutons following stimulation of the postsynaptic terminals [46-49]. In general, CB1 activation has been shown to inhibit the neurotransmission of other mediators, including glycine, acetylcholine, norepinephrine and serotonin [50], but the underpinnings of these phenomena have not been completely elucidated. Additionally, CB1 receptors have been implicated in short- and long-term synaptic depression, in relation to phasic or tonic endocannabinoid release (for a review on these topics, see [51]).

The function of CB1 receptors may vary depending on the specific interactions that they entertain with other molecular targets. For example, CB1 receptors have been found to associate with other G-protein complex receptors, such as dopamine D2, orexin Ox1, μ opioid and adenosine A2a, to form heteromeric complexes (reviewed in [52,53])

The key role of CB1 receptors as mediators of neurochemical homeostasis in the brain is maintained through a complex regulation of their expression. For example, these receptors are subjected to a rapid internalization (via clathrin-coated pits) following their binding with full agonists; on the other hand, the receptors are also recycled, with a process that requires endosomal acidification and dephosphorilation [54].

While CB2 receptors are abundantly expressed in most peripheral organs (and particularly in immune cells, where they regulate cytokine secretion and modulate cell trafficking) [55], their distribution in the brain appears to be sparse and particularly confined to microglial cells; nevertheless, recent evidence has revealed the presence of CB2 receptors in several areas of the brain [56-58]. Interestingly, a number of studies suggest that neuronal CB2 receptors may be mainly located in postsynaptic terminals [58,59]; nevertheless, the functional role of these targets in the brain remains largely elusive and awaits further characterization.

The existence of cannabinoid receptors other than CB1 and CB2 has been postulated based on ample experimental evidence [60-62]. Interestingly, a number of investigations have pointed to GPR55 as a novel putative cannabinoid receptor [63,64]; nevertheless, evidence on the specificity of this receptor for endocannabinoid is still inconclusive [65].

Endocannabinoids Both anandamide and 2-AG are derivatives of arachidonic acid, an unsaturated C20 fatty acid with 4 double bonds, which also serves as the precursor for synthesis of other eicosanoids, including prostaglandins and leukotriens. Anandamide is found in picomolar concentrations and acts as a high-affinity partial agonist for both CB1 and CB2 receptors. It is synthesized on demand by enzymatic hydrolysis of the membrane phospholipid N- arachidonoyl phosphatidylethanolamine (NAPE), a process catalyzed by several

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 5

phospholipases [66-68]. Following release and activation of CB receptors, anandamide is rapidly removed from the synaptic cleft by a carrier-mediated system [69-72] and

NIH-PA Author Manuscript NIH-PA Author Manuscriptsubsequently NIH-PA Author Manuscript hydrolyzed by the membrane enzyme fatty acid amide hydrolase (FAAH) [73-75]. FAAH serves the catabolism of other substrates, including oleoylethanolamine (OEA) and palmitoylethanolamine (PEA). Both these compounds do not activate CB1 receptors [76], although they may reduce or slow down anandamide degradation by competing with it for FAAH activity.

In comparison with anandamide, 2-AG is much more abundant (occurring in nanomolar concentrations across most tissues) and acts as a full agonist of both CB receptors. It is produced from 1,2diacylglycerol (DAG) by diacylglycerol lipase (DAGL) [77] and degraded mainly by the cytosolic serine hydrolase monoacylglycerol lipase (MAGL) [78], although other enzymes are known to contribute to this process [79].

The divergent neurochemical profiles of anandamide and 2-AG underscore their different physiological roles. Although our current understanding of the different functions entertained by each endocannabinoid is still rudimentary, the development of FAAH and MAGL inhibitors [80,81] has been instrumental to elucidate the implication of each mediator in synaptic and neurochemical regulation. While 2-AG is known as the retrograde mediator of DSI [82,83] and DSE [84-87], a number of studies suggest that anandamide may serve as an activity-dependent regulator of monoaminergic transmission [88-90]. Recent evidence points to a potential biological antagonism between anandamide and 2-AG [91,92]; on the other hand, emerging evidence points to a similar role of anandamide and 2-AG in the regulation of anxiety (albeit in relation to different receptors) and pain [93]. The development of JZL195, a potent FAAH/MAGL inhibitor, has in turn revealed that the behavioral effects of CB1 receptor agonists can be only recapitulated by the combination of both endocannabinoid-mediated functions [94].

Other lipids have been indicated as putative endocannabinoids, including 2- arachidonoylglycerylether (noladin ether) [95] and O-arachidonoylethanolamine (virodhamine) [96] (Fig. 3). Additionally, recent evidence has identified that CB receptors may be modulated by peptidic ligands, such as hemopressin and its derivatives [97,98].

EFFECTS OF CANNABIS AND CANNABINOID AGENTS ON ANXIETY

Cannabis, THC and CB1 receptor agonists The employment of cannabis for its medicinal, relaxing and mood-enhancing properties has been documented across most ancient civilizations. Originally introduced in Chinese pharmacopoeia during the third millennium BCE [99,100], cannabis became a popular remedy throughout Asia and Europe in the following centuries [99,101]. The inclusion of cannabis in the medical treatises by Dioscorides and Galen secured the herb a stable reputation in the Roman Empire and the Arabic world [101]. Until the early 20th century, the plant remained a valuable therapy for a large number of diseases [102]; however, growing concerns about the psychoactive and narcotic effects of cannabis led to a progressive restriction and ultimate ban of its usage in the United States and several European countries [100,103]. Despite its illicit status, cannabis remains one of the most popular recreational drugs, particular among adolescents and young adults, in view of its mood-enhancing and euphoriant characteristics [104-106].

Most psychological and behavioral effects of marijuana and other hemp products are induced by THC through activation of CB1 brain receptors. In fact, although THC and most synthetic cannabinoids are known to activate both CB1 and CB2 receptors, their actions on

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 6

anxiety-like behaviors and emotional regulation are efficiently countered by selective CB1 receptor antagonists, such as rimonabant (see next section) [107]. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript The studies on the psychological effects of cannabis and THC have unfolded a highly complex and often contradictory scenario, fostering a long-standing debate on the potential harms and benefits of its products. An important aspect of this discussion (particularly in consideration of its legal aspects and the potential therapeutic applications of hemp derivatives), revolves around the distinction between use and misuse of cannabis. In particular, whereas the abuse and dependence liability of cannabis is generally well- recognized [108,109], the definition of these phenomena has been heavily criticized as reflective of political agendas rather than scientific bases. For instance, the diagnosis of substance abuse, according to the criteria listed by the DSM–IV TR, is based on the manifestation of at least one of four symptoms: interference with major professional or personal obligations; intoxication in hazardous settings; substance-related legal problems; continued use in the face of persistent social or interpersonal problems [110]. The applicability of some of these standards to marijuana and other cannabis derivatives, however, has been questioned [99], also in view of their lower potential to induce physical harm in comparison with other legal substances, such as alcohol and tobacco [111].

While the controversies surrounding cannabis are far from subdued (and are often permeated and masked by conflicting ideological credos), standardized studies on cannabinoids have highlighted that the psychological and behavioral outcomes of this substance are highly variable and range from relaxation, euthymia and heightened sociability to panic, paranoid ideation and psychosis [112-116]. A corollary of this observation is that the high comorbidity rate between cannabis use disorders and psychiatric conditions [100-105] may indicate that cannabis consumption is either a concurring cause or a “self-therapeutic” strategy for anxiety and mood disorders [117-123]. The latter interpretation is supported by the observation that anxiety-spectrum disturbances and traumas in early developmental stages are a strong predictor for later cannabis use disorders [124-127]; furthermore, several lines of evidence suggest that the anxiolytic effects of THC may partially account for the high prevalence of cannabis use in patients affected by PTSD [128-131] and OCD [132]. Accordingly, recent clinical studies have shown that THC elicits therapeutic effects in OCD [133] and trichotillomania, an impulse-control disorder characterized by compulsive hair- pulling [134]. Nevertheless, prospective analyses show that cannabis use and dependence increase the risk for development of panic disorder [135], suggesting that the effect of cannabis may vary with respect to the nosological entities within the spectrum of anxiety disorders. Of note, chronic consumption of cannabis has been hypothesized to exacerbate depressive or anxious manifestations, and reduce the therapeutic efficacy of anxiolytic agents [122,136-138]; an interesting theoretical implication of this finding is that long-term exposure to cannabinoid agents may lead to profound alterations of synaptic plasticity and neurochemical homeostasis and alter the pathophysiological trajectory of anxiety and mood disorders. Thus, while cannabis may be initially used as a self-therapy for certain anxiety disorders, the prolonged exposure to this substance in vulnerable individuals may in turn alter or aggravate the clinical course of these conditions and render the patients refractory to standard treatments.

The ability of cannabis to either exacerbate or attenuate emotional reactivity is highly influenced by numerous factors, including its chemotype, as well as the influence of genetic, developmental and contextual variables. Unfortunately, little is still known about the susceptibility factors that govern the behavioral outcomes of cannabis in patients affected by anxiety-spectrum disorders. Indeed, several components have been shown to play a role in this link, including genetic background, age, gender, environmental stress and concurrent

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 7

use of other drugs; a detailed analysis of these determinants is outside the scope of the present work, but the interested reader should refer to [139]. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript Aside from the influence of vulnerability factors, the available evidence indicates that cannabis, THC and other CB1 receptor agonists exercise a bidirectional influence on anxiety responses as a function of the dosage. The majority of users report that consumption of modest amounts of cannabis and CB1 receptor agonists results in euphoria, relaxation, heightened perception, sociability and creativity, moderate to high doses have been reported to elicit phobia, agitation, panic, dysphoria, psychotic manifestations and cognitive impairments [112-116,124,140-143]. In line with these premises, early studies showed a robust anxiolytic efficacy of low-dose nabilone in comparison with placebo [144,145]. Additionally, the few available reports on the clinical outcomes of recreational cannabinoids show that a moderate consumption of “Spice” blends is generally associated with euphoria and disinhibition [146], but the abuse of these substances is conducive to high levels of anxiety, panic, paranoid ideation and mood disturbances [147-151].

The biphasic effects of cannabinoids on anxiety-related responses have been extensively documented in rodents. In agreement with human evidence, preclinical studies have elucidated that the acute administration of low doses of CB1 receptor agonists elicits anxiolytic-like in approach/avoidance tasks [152-156]; conversely, high concentrations of the same compounds are generally associated with the opposite outcomes [157-162] (for complete reviews of the topic, see [163,164])

The bidirectional action of CB1 receptors on anxiety responses may be related to the modulatory role of these targets on GABA and glutamate release across amygdala and other forebrain areas [41,165,166]. As these two major neurotransmitters affect anxiety in an opposite fashion, different doses of cannabinoids and synthetic CB1 receptor agonists may indeed produce highly divergent effects, in relation to their ability to affect the homeostasis and the balance of GABA and glutamate (for a review on these issues, see [163]). Furthermore, CB1 receptors have been shown to play critical roles in the regulation of most neurochemical substrates of anxiety, including the neurotransmitters serotonin, norepinephrine and acetylcholine, as well as stress hormones, colecystokynin and opioid peptides [50,163].

In line with this concept, the infusion in the periaqueductal grey of arachidonyl-2- chloroethylamide (ACEA), an anandamide synthetic analog with high CB1 receptor selectivity, elicited anxiolytic-like effects in rats in an elevated plus maze, with a bell-shaped dose-response curve [167], the highest doses being associated to no significant behavioral change. Novel categories of compounds have been patented for potential efficacy as selective CB1 receptor modulators, including sulfonyl-benzamides [168] and tetrasubstituted imidazole derivatives [169]. To the best of our knowledge, however, no findings on the action of these compounds in anxiety regulation have been reported to date.

CB1 receptor antagonists/inverse agonists

The cannabinoid CB1 receptor antagonists/inverse agonist rimonabant was introduced into clinical practice by Sanofi-Aventis in 2006 as a treatment for obesity [170] and smoking cessation [171]. The majority of preclinical studies found that these compounds are anxiogenic at high doses [158,159,172,173] and ineffective at low doses [174,175]. The anxiogenic properties of CB1 antagonists, were unequivocally confirmed by clinical data on the psychiatric side effects of rimonabant. The significant increase in anxiety, depression and suicidality in patients under treatment with rimonabant [176-179], in particular, led to the withdrawal of the drug from the European market in October, 2008. As a consequence, several pharmaceutical companies announced the interruption of their clinical research on

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 8

CB1 receptor antagonists, including taranabant (from Merck) and otenabant (from Pfizer), both in Phase 3 of development. Some of the anxiogenic properties of rimonabant and

NIH-PA Author Manuscript NIH-PA Author Manuscriptanalogs NIH-PA Author Manuscript have been speculated to be due to their activity as inverse agonists; as a result, the therapeutic use of newly-developed neutral CB1 antagonists has been proposed, with the hypothesis that these compounds would not elicit the untoward psychological effects observed with rimonabant and its analogs [180,181]; this idea is supported by recent findings, showing that unlike CB1 receptor inverse agonists, the neutral antagonists of this targets fail to facilitate the acquisition or consolidation of fear [182].

CB2 receptor ligands

Few studies have actually evaluated the role of CB2 receptor in anxiety and stress response. While this receptor was posited to be mainly expressed mainly in immune cells and peripheral areas, its identification in the brain under pathological conditions, such as Alzheimer’s disease, multiple sclerosis and amyotrophic lateral sclerosis spinal cord [183-185], led to a number of studies aimed at the assessment of its potential role in brain function and behavioral regulation. Some of these investigations indicated that the suppression of CB2 receptor in the brain, through intracerebroventricular injection of antisense nucleotide sequences, elicited anxiolytic effects in rodents [186]. In contrast, Garcia-Gutierrez and Manzanares [187] recently described that the overexpression of CB2 receptors reduced anxiogenic-related behaviors in the light-dark box and elevated plus maze. These premises point to the possibility that CB2 receptor ligands may also play a role in the modulation of anxiety disorders. This hypothesis, however, awaits further examination with proper pharmacological tools.

CBD Several studies suggest that THC and CBD may exert opposite actions on brain function and psychopathology [188], possibly in relation to the action of CBD as a potent CB1 receptor antagonist/inverse agonist [21] (see above). Several lines of preclinical work have shown that CBD reduces the effects of THC on several behavioral functions [189-191]. In line with these data, CBD has been found to reduce the anxiety and improve the sensation of well being induced by an acute, high THC dose in healthy volunteers [192].

In contrast with these data, a number of studies have shown that CBD pretreatment potentiated the behavioral effects induced by THC [193-195]. These actions may signify the ability of CBD to inhibit cytochrome P450-mediated drug metabolism [196,197], which may increase THC blood and brain concentrations [193,195].

Notably, the behavioral outcomes of CBD do not appear to be only due to potential pharmacodynamic/pharmacokynetic competition with THC; indeed, recent studies have shown that CBD exerts inherent anxiolytic effects, both in rodent models [157,198-201] and, more recently, in patients affected by social phobia [202,203]. The anxiolytic action of CBD may be linked to 5-HT1A receptor, but not through benzodiazepine receptors [204]. Of note, the anxiolytic action of CBD also appears to be bidirectional, as only low to moderate doses, but not high doses, have been associated with exert anxiolytic effects [200,205].

The anxiolytic action of CBD do not appear to be mediated by benzodiazepine receptors [204], but rather by 5-HT1A serotonin receptors in the bed nucleus of the stria terminalis [206], a critical component of the amygdaloid complex involved in the regulation of stress response.

Accordingly, CBD has been shown to reduce amygdalar responses to fearful stimuli [207]; this mechanism may be essential for the anxiolytic effects of this compound in social phobia [203]. Furthermore, CBD has been shown to elicit antipanic effects through the activation of

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 9

5-HT1A receptors in the dorsal periaqueductal gray, a critical area for the modulation of emotional reactivity to stress [208,209]. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript Endocannabinoid transport blockers The systemic administration of the endocannabinoid transport blocker AM404 (Fig. 4) was shown to elicit anxiolytic-like behaviors in the elevated plus maze and defensive withdrawal in adult rats, as well as an attenuation of ultrasonic vocalizations in rat pups [175]. The same compound was shown to attenuate marble burying (a paradigm for compulsivity testing) in mice, suggesting that this compound may have some potential efficacy for OCD [206]. Interestingly, the anxiolytic effects of AM404 were shown to be contributed by both CB1 and 5-HT1A receptors [152,210], in a fashion similar to the potent CB1 receptor agonist CP 55,940 [160]. Additionally, AM404 has been suggested to act as a FAAH inhibitor [211], although evidence in this respect is controversial [72]. Indeed, despite the identification of potential candidate endocannabinoid binding sites [212], no final evidence is currently available on the existence and/or molecular identity of the endocannabinoid transporter.

Although the possibility of targeting the endocannabinoid carrier for the development of anxiolytic compounds is appealing and has been targeted by a patent proposing these compounds as a pharmacological support for psychotherapy [213], the elusive molecular identity of the transporter itself has greatly limited the studies. Furthermore, preliminary data indicate that AM404 elicits reward in animals and is self-administered by squirrel monkeys [175,214], raising the possibility that endocannabinoid transport blockers may be addictive.

FAAH inhibitors The prototypical FAAH inhibitor URB597 (Fig. 4) has been shown to reduce anxiety-like behaviors in rats, in a rimonabant-sensitive fashion [155,163,215-217]. In addition to its anxiolytic-like properties, URB597 was found to exert also antidepressant-like effects in several animal models with high face and predictive validity, such as the forced swim, tail suspension and chronic mild stress paradigms [89,210,216,218]. The anxiolytic action of FAAH inhibitors has been suggested to depend on the enhancement of anandamide in the dorsolateral periaqueductal gray [219]; interestingly, however, only low doses of URB597 in the prefrontal cortex were found to elicit anxiolytic-like effects, through CB1 receptor activation. However, higher doses ceased to elicit anxiolysis, in view of their interaction with TPRV1 vanilloid receptors [220]. Furthermore, the anxiolytic and antidepressant actions of FAAH inhibitors were observed only under conditions of high environmental aversiveness, but not under normal conditions [163,218,221]. Importantly, the psychotropic effects of FAAH inhibitors are partially distinct from those associated with cannabinoids, in that they appear to fail to reproduce the hedonic and interoceptive states produced by CB receptor agonists [89] and to induce self-administration in squirrel monkeys [222]. Taken together, these data suggest that FAAH inhibitors may be promising tools in the therapy of anxiety and mood disorders with a safer profile than cannabinoid direct agonists. This idea has been recently endorsed by several authors in recent articles and patents, featuring novel categories of highly selective and potent FAAH inhibitors [223-225] [226]. However, it should be noted that recent data have recently shown that URB597 induce a number of side effects in rats, including social withdrawal, working memory deficits [227] and impairments in auditory discrimination and reversal of olfactory discrimination [228].

MAGL inhibitors The role of 2-AG in emotional regulation has been difficult to ascertain until the recent development of highly selective monoacylglycerol lipase (MAGL) inhibitors [35,223]. Several lines of evidence have suggested that 2-AG plays a pivotal role in the

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 10

pathophysiology of anxiety and defensive behaviors. The prototypical MAGL inhibitor, JZL184 (Fig. 4), has been shown to enhance the levels of 2-AG, but not anandamide; this

NIH-PA Author Manuscript NIH-PA Author Manuscripteffect NIH-PA Author Manuscript is due to its extremely high selectivity for MAGL over FAAH and other brain serine hydrolases. Recent evidence has shown that this compound exerts anxiolytic-like effects in the elevated plus maze and in marble buyring, at doses that do not affect locomotor activity [93,229,230]. Similarly to the effects described for FAAH inhibitors (see above), the anxiolytic effects of this compound were observed in highly aversive (or anxiogenic) contextual settings [229]. The neurobiological role of 2-AG in anxiety is still poorly understood, although several studies have shown that environmental stressors alter its biosynthesis and degradation in key brain structures controlling emotional regulation, including periaqueductal grey, amygdala and hippocampus [231,232]. Interestingly, recent evidence has shown that the anxiolytic properties of JZL184 appear to be mediated by CB2, rather than CB1 receptors [93], pointing to a potential implication of this receptor in the role of 2-AG in anxiety regulation.

CURRENT AND FUTURE DEVELOPMENTS In light of the limitations of our current pharmacological armamentarium for anxiety disorders, the ability of cannabinoids to modulate emotional responses is extremely attractive for the development of novel anxiolytic agents [217]. At the same time, great concern arises from the protean role of cannabinoids on the regulation of these responses, as well as their misuse liability and other side effects. The identification of operational strategies for the employment of cannabinoids in the therapy of anxiety disorders is therefore a fundamental goal in psychiatry research.

As outlined above, clinical evidence strongly suggests that acute administration of low doses of CB1 receptor agonists results in anxiolytic effects, while excessive activation of these targets elicits opposite outcomes, following a reverse U-shaped dose-response pattern. Hence, a primary strategy to harness the anxiolytic properties of cannabinoids could consist in the employment of partial, low-affinity CB1 agonists, which may ensure a relatively high therapeutic index and the stabilization of the activation of this target within a range associated with mood enhancement and/or anxiolysis. This idea is indirectly supported by the mirroring observation that anecdotal reports on highly potent, high-affinity synthetic cannabinoids (such as those contained in “Spice” blends) trigger greater psychoactive effects than the partial CB agonist THC [26]. This concept indicates a potential evolution in the search for direct CB agonists, in sharp contrast with the previous trend aimed at the identification of high-affinity CB receptor activators.

An alternative strategy to achieve a similar therapeutic goal may lie in the combination of CB1 receptor agonists with low dosages of antagonists (preferably neutral, in order to avoid potential side effects linked to CB1 inverse agonism); this intriguing approach, which has been indicated in a recent patent [233], is based on the likely mechanism of action of Sativex®, a cannabinoid mouth spray containing THC and CBD (in a ratio of 1.08:1) and marketed for the treatment of neuropathic pain, spasticity and overactive bladder, in consideration of the action of CBD as a CB1 receptor antagonist. However, recent preliminary clinical studies have shown that this formulation did not significantly reduce anxiety (in fact, it was reported to induce a mild, yet not significant increase of this symptom) [234,235], and that CBD did not appear to elicit a significant opposition to the effect of dronabinol [235], plausibly indicating that a higher concentration of this ingredient (or lower relative amount of THC) may be necessary to elicit anxiolytic effects.

A third, highly promising avenue for the development of cannabinoid-based anxiolytic therapies may be afforded by FAAH inhibitors. Unlike endocannabinoid transport blockers

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 11

and direct CB receptor agonists, these compounds exhibit a number of highly desirable properties for anxiolytic agents: first, they appear to maintain their anxiolytic and

NIH-PA Author Manuscript NIH-PA Author Manuscriptantidepressant NIH-PA Author Manuscript effect not only under conditions of acute administration, but also following long-term treatment [93,210]; second, they appear to elicit their effects only in conditions of highly aversive environmental circumstances (i.e., similar to those that would in fact require an anxiolytic treatment); third, they have no apparent addiction liability [89,222]. The neurobiological bases of this phenomenon are not completely understood, and may be related to the involvement of other FAAH substrates, such as OEA or PEA; however, recent investigations suggest that the lack of 2-AG enhancement ensuing FAAH inactivation may contribute to the lack of reinforcing properties of URB597 [236], potentially suggesting a different role of anandamide and 2-AG in the modulation of reward; this idea is actually consistent with the recent finding that 2-AG is induces self-administration in monkeys [237].

A key problem concerning the potential application of cannabinoid-related agents and cannabinoids is the relatively little information about their long-term effects following chronic administration. Indeed, the subjective effects of cannabis have been shown to be typically different in chronic users as compared to occasional marijuana smokers [238,239]. Prolonged consumption of cannabis has been shown to induce affective sequelae, including alexithymia and avolition [113,240-242]. Interestingly, tolerance has been shown to the effects of THC [243,244], while no information is available on endocannabinoid-related agents. Long-term administration of cannabinoids has been shown to result in a number of neuroplastic adaptive processes, including CB receptor down-regulation [245,246]. Some of these phenomena may indeed be critical in shaping the different emotional responsiveness to cannabis throughout life and reflect a potential pathophysiological loop which may compound the severity of pre-existing anxiety and affective disorders.

Finally, another important step for the employment of cannabinoid-based anxiolytic therapies will be the analysis of the vulnerability factors implicated in the differential responses and long-term sequelae induced by cannabis consumption. For example, numerous meta-analyses and longitudinal studies have established that cannabis consumption in adolescence is conducive to an increased risk for psychotic disorders [247-250]. This association is particularly significant in the presence of other genetic factors, such as the Val108Met allelic variant of the gene encoding Catechol-O- methyltransferase (COMT) [251,252], one of the main enzymes for the degradation of the neurotransmitter dopamine. Interestingly, it has been shown that the synergistic effect of COMT haplotype and cannabis in adolescence is more robust in conjunction with predisposing environmental variables, such as the exposure to urbanicity and psychosocial stress [253]. Another gene that may modulate the behavioral responsiveness to cannabinoids is Nrg1, which encodes for the synaptic protein neuregulin 1. Indeed, the heterozygous deletion of this gene ablates the development of tolerance to the anxiogenic effects of CB receptor agonists [254,255]. These findings suggest that the employment of a pharmacogenetic approach may be a critical screening instrument to identify which patients may be treated with cannabis for medical purposes without risks of neuropsychiatric side effects. Notably, the role of genes in the mental sequelae of cannabis may also be contributed by epigenetic factors, in consideration of the recent finding that THC induces expression of histone deacetylase 3 [256].

While studies on the biological determinants of different responses to cannabis are still at their preliminary stages, advances in this area may be essential to allow a personalized approach for the employment of cannabinoid-based therapies in anxiety and mood disorders.

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 12

Acknowledgments

The present work was supported by the National Institute of Health grant R21HD070611 and the USC Zumberge NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript Individual Research Grant (to MB).

REFERENCES 1. Nutt D, Allgulander C, Lecrubier Y, Peters T, Wittchen U. Establishing non-inferiority in treatment trials in psychiatry: guidelines from an Expert Consensus Meeting. J Psychopharmacol. 2008; 22(4): 409–16. [PubMed: 18635721] 2. Sugiura T, Waku K. 2-Arachidonoylglycerol and the cannabinoid receptors. Chem Phys Lipids. 2000; 108(1-2):89–106. [PubMed: 11106784] 3. Hillig KW, Mahlberg PG. A chemotaxonomic analysis of cannabinoid variation in Cannabis (Cannabaceae). Am J Bot. 2004; 91(6):966–75. [PubMed: 21653452] 4. ElSohly MA, Slade D. Chemical constituents of marijuana: the complex mixture of natural cannabinoids. Life sciences. 2005; 78(5):539–48. [PubMed: 16199061] 5. Brenneisen, R. Chemistry and Analysis of Phytocannabinoids and Other Cannabis Constituents. In: ElSohly, MA., editor. Marijuana and the Cannabinoids. Humana Press; 2007. p. 17-49. 6. ElSohly, MA.; Stanford, DF.; Murphy, TP. Chemical Fingerprinting of Cannabis as a Means of Source Identification. In: ElSohly, MA., editor. Marijuana and the Cannabinoids. Humana Press; 2007. p. 51-66. 7. Miller Coyle H, Palmbach T, Juliano N, Ladd C, Lee HC. An overview of DNA methods for the identification and individualization of marijuana. Croatian medical journal. 2003; 44(3):315–21. [PubMed: 12808725] 8. Brown H. Possible anticholinesterase-like effects of trans(−) 8 and - 9 tetrahydrocannabinol as observed in the general motor activity of mice. Psychopharmacologia. 1972; 27(2):111–6. [PubMed: 4638205] 9. Yoshimura H, Fujiwara M, Ueki S. Biochemical correlates in mouse-killing behavior of the rat: brain acetylcholine and acetylcholinesterase after administration of delta9-tetrahydrocannabinol. Brain research. 1974; 81(3):567–70. [PubMed: 4474050] 10. Mishima K, Egashira N, Matsumoto Y, Iwasaki K, Fujiwara M. Involvement of reduced acetylcholine release in Delta9-tetrahydrocannabinol-induced impairment of spatial memory in the 8-arm radial maze. Life sciences. 2002; 72(4-5):397–407. [PubMed: 12467880] 11. Zuardi AW. Cannabidiol: from an inactive cannabinoid to a drug with wide spectrum of action. Rev Bras Psiquiatr. 2008; 30(3):271–80. [PubMed: 18833429] 12. de Meijer EPM, Kamp HJ, Eeuwijk FA. Characterisation of Cannabis accessions with regard to cannabinoid content in relation to other plant characters. Euphytica. 1992; 62(3):187–200. 13. Fadda P, Robinson L, Fratta W, Pertwee RG, Riedel G. Differential effects of THC- or CBD-rich cannabis extracts on working memory in rats. Neuropharmacology. 2004; 47(8):1170–9. [PubMed: 15567426] 14. Ryan D, Drysdale AJ, Pertwee RG, Platt B. Differential effects of cannabis extracts and pure plant cannabinoids on hippocampal neurones and glia. Neurosci Lett. 2006; 408(3):236–41. [PubMed: 16997463] 15. Starks, M. Marijuana chemistry: genetics, processing & potency. Ronin Publishing, Inc.; Oakland, USA: 1990. 16. Showalter VM, Compton DR, Martin BR, Abood ME. Evaluation of binding in a transfected cell line expressing a peripheral cannabinoid receptor (CB2): identification of cannabinoid receptor subtype selective ligands. J Pharmacol Exp Ther. 1996; 278(3):989–99. [PubMed: 8819477] 17. Thomas BF, Gilliam AF, Burch DF, Roche MJ, Seltzman HH. Comparative receptor binding analyses of cannabinoid agonists and antagonists. J Pharmacol Exp Ther. 1998; 285(1):285–92. [PubMed: 9536023] 18. Pertwee RG. Pharmacology of cannabinoid receptor ligands. Curr Med Chem. 1999; 6(8):635–64. [PubMed: 10469884]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 13

19. Bisogno T, Hanus L, De Petrocellis L, Tchilibon S, Ponde DE, Brandi I, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001; 134(4):845–52. [PubMed: NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 11606325] 20. Thomas A, Ross RA, Saha B, Mahadevan A, Razdan RK, Pertwee RG. 6″-Azidohex-2″-yne- cannabidiol: a potential neutral, competitive cannabinoid CB1 receptor antagonist. Eur J Pharmacol. 2004; 487(1-3):213–21. [PubMed: 15033394] 21. Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007; 150(5):613–23. [PubMed: 17245363] 22. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9- tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol. 2008; 153(2):199–215. [PubMed: 17828291] 23. Fisar Z. Phytocannabinoids and endocannabinoids. Curr Drug Abuse Rev. 2009; 2(1):51–75. [PubMed: 19630737] 24. Cascio MG, Gauson LA, Stevenson LA, Ross RA, Pertwee RG. Evidence that the plant cannabinoid cannabigerol is a highly potent alpha2-adrenoceptor agonist and moderately potent 5HT1A receptor antagonist. Br J Pharmacol. 2010; 159(1):129–41. [PubMed: 20002104] 25. Davis MP. Oral nabilone capsules in the treatment of chemotherapy-induced nausea and vomiting and pain. Expert Opin Investig Drugs. 2008; 17(1):85–95. 26. Atwood BK, Huffman J, Straiker A, Mackie K. JWH018, a common constituent of ‘Spice’ herbal blends, is a potent and efficacious cannabinoid CB receptor agonist. Br J Pharmacol. 2010; 160(3): 585–93. [PubMed: 20100276] 27. Atwood BK, Lee D, Straiker A, Widlanski TS, Mackie K. CP47,497-C8 and JWH073, commonly found in ‘Spice’ herbal blends, are potent and efficacious CB(1) cannabinoid receptor agonists. Eur J Pharmacol. 2011; 659(2-3):139–45. [PubMed: 21333643] 28. Gaoni Y, Mecbonlam R. Isolation, Structure, and Partial Synthesis of an Active Constituent of Hashish. J Amer Chem Soc. 1964; 86:1646–7. 29. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990; 346(6284):561–4. [PubMed: 2165569] 30. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993; 365(6441):61–5. [PubMed: 7689702] 31. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992; 258(5090): 1946–9. [PubMed: 1470919] 32. Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995; 50(1):83–90. [PubMed: 7605349] 33. Sugiura T, Kondo S, Sukagawa A, Nakane S, Shinoda A, Itoh K, et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995; 215(1):89–97. [PubMed: 7575630] 34. Howlett AC, Qualy JM, Khachatrian LL. Involvement of Gi in the inhibition of adenylate cyclase by cannabimimetic drugs. Mol Pharmacol. 1986; 29(3):307–13. [PubMed: 2869405] 35. Adamec R. Transmitter systems involved in neural plasticity underlying increased anxiety and defense--implications for understanding anxiety following traumatic stress. Neurosci Biobehav Rev. 1997; 21(6):755–65. [PubMed: 9415900] 36. Demuth DG, Molleman A. Cannabinoid signalling. Life sciences. 2006; 78(6):549–63. [PubMed: 16109430] 37. Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, de Costa BR, et al. Cannabinoid receptor localization in brain. Proc Natl Acad Sci U S A. 1990; 87(5):1932–6. [PubMed: 2308954] 38. Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC. Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci. 1991; 11(2):563–83. [PubMed: 1992016]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 14

39. Charney DS, Deutch A. A functional neuroanatomy of anxiety and fear: implications for the pathophysiology and treatment of anxiety disorders. Crit Rev Neurobiol. 1996; 10(3-4):419–46. [PubMed: 8978989] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 40. Glass M, Dragunow M, Faull RL. Cannabinoid receptors in the human brain: a detailed anatomical and quantitative autoradiographic study in the fetal, neonatal and adult human brain. Neuroscience. 1997; 77(2):299–318. [PubMed: 9472392] 41. Katona I, Rancz EA, Acsady L, Ledent C, Mackie K, Hajos N, et al. Distribution of CB1 cannabinoid receptors in the amygdala and their role in the control of GABAergic transmission. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2001; 21(23): 9506–18. [PubMed: 11717385] 42. Freund TF, Katona I, Piomelli D. Role of endogenous cannabinoids in synaptic signaling. Physiol Rev. 2003; 83(3):1017–66. [PubMed: 12843414] 43. Mackie K. Distribution of cannabinoid receptors in the central and peripheral nervous system. Handb Exp Pharmacol. 2005; (168):299–325. [PubMed: 16596779] 44. Tsou K, Brown S, Sanudo-Pena MC, Mackie K, Walker JM. Immunohistochemical distribution of cannabinoid CB1 receptors in the rat central nervous system. Neuroscience. 1998; 83(2):393–411. [PubMed: 9460749] 45. Salio C, Fischer J, Franzoni MF, Conrath M. Pre- and postsynaptic localizations of the CB1 cannabinoid receptor in the dorsal horn of the rat spinal cord. Neuroscience. 2002; 110(4):755–64. [PubMed: 11934482] 46. Morishita W, Alger BE. Evidence for endogenous excitatory amino acids as mediators in DSI of GABA(A)ergic transmission in hippocampal CA1. J Neurophysiol. 1999; 82(5):2556–64. [PubMed: 10561426] 47. Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001; 410(6828):588–92. [PubMed: 11279497] 48. Ohno-Shosaku T, Maejima T, Kano M. Endogenous cannabinoids mediate retrograde signals from depolarized postsynaptic neurons to presynaptic terminals. Neuron. 2001; 29(3):729–38. [PubMed: 11301031] 49. Varma N, Carlson GC, Ledent C, Alger BE. Metabotropic glutamate receptors drive the endocannabinoid system in hippocampus. J Neurosci. 2001; 21(24):RC188. [PubMed: 11734603] 50. Szabo B, Schlicker E. Effects of cannabinoids on neurotransmission. Handb Exp Pharmacol. 2005; (168):327–65. [PubMed: 16596780] 51. Lovinger DM. Endocannabinoid Signaling in Neural Plasticity. Pharmacology of Neurotransmitter Release. 2008; 184:435–77. 52. Pertwee RG, Howlett AC, Abood ME, Alexander SP, Di Marzo V, Elphick MR, et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB and CB. Pharmacological reviews. 2010; 62(4):588–631. [PubMed: 21079038] 53. Hudson S, Ramsey J, King L, Timbers S, Maynard S, Dargan PI, et al. Use of high-resolution accurate mass spectrometry to detect reported and previously unreported cannabinomimetics in “herbal high” products. J Anal Toxicol. 2010; 34(5):252–60. [PubMed: 20529459] 54. Hsieh C, Brown S, Derleth C, Mackie K. Internalization and Recycling of the CB1 Cannabinoid Receptor. Journal of Neurochemistry. 1999; 73(2):493–501. [PubMed: 10428044] 55. Walter L, Stella N. Cannabinoids and neuroinflammation. Br J Pharmacol. 2004; 141(5):775–85. [PubMed: 14757702] 56. Van Sickle MD, Duncan M, Kingsley PJ, Mouihate A, Urbani P, Mackie K, et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science. 2005; 310(5746):329–32. [PubMed: 16224028] 57. Gong JP, Onaivi ES, Ishiguro H, Liu QR, Tagliaferro PA, Brusco A, et al. Cannabinoid CB2 receptors: immunohistochemical localization in rat brain. Brain research. 2006; 1071(1):10–23. [PubMed: 16472786] 58. Onaivi ES. Neuropsychobiological evidence for the functional presence and expression of cannabinoid CB2 receptors in the brain. Neuropsychobiology. 2006; 54(4):231–46. [PubMed: 17356307]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 15

59. Brusco A, Tagliaferro P, Saez T, Onaivi ES. Postsynaptic localization of CB2 cannabinoid receptors in the rat hippocampus. Synapse. 2008; 62(12):944–9. [PubMed: 18798269]

NIH-PA Author Manuscript NIH-PA Author Manuscript60. Breivogel NIH-PA Author Manuscript CS, Griffin G, Di Marzo V, Martin BR. Evidence for a new G protein-coupled cannabinoid receptor in mouse brain. Mol Pharmacol. 2001; 60(1):155–63. [PubMed: 11408610] 61. O’Sullivan SE, Kendall DA, Randall MD. Heterogeneity in the mechanisms of vasorelaxation to anandamide in resistance and conduit rat mesenteric arteries. Br J Pharmacol. 2004; 142(3):435– 42. [PubMed: 15148250] 62. Begg M, Pacher P, Batkai S, Osei-Hyiaman D, Offertaler L, Mo FM, et al. Evidence for novel cannabinoid receptors. Pharmacol Ther. 2005; 106(2):133–45. [PubMed: 15866316] 63. Sjögren, S.; Ryberg, E.; Lindblom, A.; Larsson, N.; Åstrand, A.; Hjorth, S., et al. A new receptor for cannabinoid ligands; Symposium on the Cannabinoids; International Cannabinoid Research Society, Burlington, Vermont, USA. 2005; 64. Lauckner JE, Jensen JB, Chen HY, Lu HC, Hille B, Mackie K. GPR55 is a cannabinoid receptor that increases intracellular calcium and inhibits M current. Proc Natl Acad Sci U S A. 2008; 105(7):2699–704. [PubMed: 18263732] 65. Brown AJ, Robin Hiley C. Is GPR55 an anandamide receptor? Vitam Horm. 2009; 81:111–37. [PubMed: 19647110] 66. Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N. Molecular characterization of a phospholipase D generating anandamide and its congeners. J Biol Chem. 2004; 279(7):5298–305. [PubMed: 14634025] 67. Sun YX, Tsuboi K, Okamoto Y, Tonai T, Murakami M, Kudo I, et al. Biosynthesis of anandamide and N-palmitoylethanolamine by sequential actions of phospholipase A2 and lysophospholipase D. Biochem J. 2004; 380(Pt 3):749–56. [PubMed: 14998370] 68. Liu C, Walker JM. Effects of a cannabinoid agonist on spinal nociceptive neurons in a rodent model of neuropathic pain. J Neurophysiol. 2006; 96(6):2984–94. [PubMed: 16943316] 69. Di Marzo V, Fontana A, Cadas H, Schinelli S, Cimino G, Schwartz JC, et al. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature. 1994; 372(6507): 686–91. [PubMed: 7990962] 70. Beltramo M, Stella N, Calignano A, Lin SY, Makriyannis A, Piomelli D. Functional role of high- affinity anandamide transport, as revealed by selective inhibition. Science. 1997; 277(5329):1094– 7. [PubMed: 9262477] 71. Hillard CJ, Campbell WB. Biochemistry and pharmacology of arachidonylethanolamide, a putative endogenous cannabinoid. J Lipid Res. 1997; 38(12):2383–98. [PubMed: 9458263] 72. Fegley D, Kathuria S, Mercier R, Li C, Goutopoulos A, Makriyannis A, et al. Anandamide transport is independent of fatty-acid amide hydrolase activity and is blocked by the hydrolysis- resistant inhibitor AM1172. Proc Natl Acad Sci U S A. 2004; 101(23):8756–61. [PubMed: 15138300] 73. Hillard CJ, Wilkison DM, Edgemond WS, Campbell WB. Characterization of the kinetics and distribution of N-arachidonylethanolamine (anandamide) hydrolysis by rat brain. Biochim Biophys Acta. 1995; 1257(3):249–56. [PubMed: 7647100] 74. Ueda N, Yamamoto K, Yamamoto S, Tokunaga T, Shirakawa E, Shinkai H, et al. Lipoxygenase- catalyzed oxygenation of arachidonylethanolamide, a cannabinoid receptor agonist. Biochim Biophys Acta. 1995; 1254(2):127–34. [PubMed: 7827116] 75. Cravatt BF, Giang DK, Mayfield SP, Boger DL, Lerner RA, Gilula NB. Molecular characterization of an enzyme that degrades neuromodulatory fatty-acid amides. Nature. 1996; 384(6604):83–7. [PubMed: 8900284] 76. LoVerme J, La Rana G, Russo R, Calignano A, Piomelli D. The search for the palmitoylethanolamide receptor. Life sciences. 2005; 77(14):1685–98. [PubMed: 15963531] 77. Bisogno T, Howell F, Williams G, Minassi A, Cascio MG, Ligresti A, et al. Cloning of the first sn1-DAG lipases points to the spatial and temporal regulation of endocannabinoid signaling in the brain. J Cell Biol. 2003; 163(3):463–8. [PubMed: 14610053] 78. Dinh TP, Freund TF, Piomelli D. A role for monoglyceride lipase in 2-arachidonoylglycerol inactivation. Chem Phys Lipids. 2002; 121(1-2):149–58. [PubMed: 12505697]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 16

79. Kozak KR, Prusakiewicz JJ, Marnett LJ. Oxidative metabolism of endocannabinoids by COX-2. Curr Pharm Des. 2004; 10(6):659–67. [PubMed: 14965328]

NIH-PA Author Manuscript NIH-PA Author Manuscript80. Piomelli NIH-PA Author Manuscript D. The endocannabinoid system: a drug discovery perspective. Curr Opin Investig Drugs. 2005; 6(7):672–9. 81. Hohmann AG. Inhibitors of monoacylglycerol lipase as novel analgesics. Br J Pharmacol. 2007; 150(6):673–5. [PubMed: 17293886] 82. Kim J, Alger BE. Inhibition of cyclooxygenase-2 potentiates retrograde endocannabinoid effects in hippocampus. Nat Neurosci. 2004; 7(7):697–8. [PubMed: 15184902] 83. Makara JK, Mor M, Fegley D, Szabo SI, Kathuria S, Astarita G, et al. Selective inhibition of 2-AG hydrolysis enhances endocannabinoid signaling in hippocampus. Nat Neurosci. 2005; 8(9):1139– 41. [PubMed: 16116451] 84. Gerdeman GL, Ronesi J, Lovinger DM. Postsynaptic endocannabinoid release is critical to long- term depression in the striatum. Nat Neurosci. 2002; 5(5):446–51. [PubMed: 11976704] 85. Robbe D, Alonso G, Chaumont S, Bockaert J, Manzoni OJ. Role of p/q-Ca2+ channels in metabotropic glutamate receptor 2/3-dependent presynaptic long-term depression at nucleus accumbens synapses. J Neurosci. 2002; 22(11):4346–56. [PubMed: 12040040] 86. Sjostrom PJ, Turrigiano GG, Nelson SB. Neocortical LTD via coincident activation of presynaptic NMDA and cannabinoid receptors. Neuron. 2003; 39(4):641–54. [PubMed: 12925278] 87. Jung KM, Mangieri R, Stapleton C, Kim J, Fegley D, Wallace M, et al. Stimulation of endocannabinoid formation in brain slice cultures through activation of group I metabotropic glutamate receptors. Mol Pharmacol. 2005; 68(5):1196–202. [PubMed: 16051747] 88. Giuffrida A, Parsons LH, Kerr TM, Rodriguez de Fonseca F, Navarro M, Piomelli D. Dopamine activation of endogenous cannabinoid signaling in dorsal striatum. Nat Neurosci. 1999; 2(4):358– 63. [PubMed: 10204543] 89. Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, et al. Antidepressant- like activity and modulation of brain monoaminergic transmission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A. 2005; 102(51):18620–5. [PubMed: 16352709] 90. Muntoni AL, Pillolla G, Melis M, Perra S, Gessa GL, Pistis M. Cannabinoids modulate spontaneous neuronal activity and evoked inhibition of locus coeruleus noradrenergic neurons. Eur J Neurosci. 2006; 23(9):2385–94. [PubMed: 16706846] 91. Di Marzo V, Cristino L. Why endocannabinoids are not all alike. Nat Neurosci. 2008; 11(2):124–6. [PubMed: 18227793] 92. Maccarrone M, Rossi S, Bari M, De Chiara V, Fezza F, Musella A, et al. Anandamide inhibits metabolism and physiological actions of 2-arachidonoylglycerol in the striatum. Nat Neurosci. 2008; 11(2):152–9. [PubMed: 18204441] 93. Busquets-Garcia A, Puighermanal E, Pastor A, de la Torre R, Maldonado R, Ozaita A. Differential Role of Anandamide and 2-Arachidonoylglycerol in Memory and Anxiety-like Responses. Biological psychiatry. 2011; 70(5):479–86. [PubMed: 21684528] 94. Long JZ, Li W, Booker L, Burston JJ, Kinsey SG, Schlosburg JE, et al. Selective blockade of 2- arachidonoylglycerol hydrolysis produces cannabinoid behavioral effects. Nat Chem Biol. 2009; 5(1):37–44. [PubMed: 19029917] 95. Hanus L, Abu-Lafi S, Fride E, Breuer A, Vogel Z, Shalev DE, et al. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci U S A. 2001; 98(7): 3662–5. [PubMed: 11259648] 96. Porter AC, Sauer JM, Knierman MD, Becker GW, Berna MJ, Bao J, et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther. 2002; 301(3):1020–4. [PubMed: 12023533] 97. Heimann AS, Gomes I, Dale CS, Pagano RL, Gupta A, de Souza LL, et al. Hemopressin is an inverse agonist of CB1 cannabinoid receptors. Proc Natl Acad Sci U S A. 2007; 104(51):20588– 93. [PubMed: 18077343] 98. Gomes I, Grushko JS, Golebiewska U, Hoogendoorn S, Gupta A, Heimann AS, et al. Novel endogenous peptide agonists of cannabinoid receptors. FASEB J. 2009; 23(9):3020–9. [PubMed: 19380512]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 17

99. Earlywine, M. Understanding marijuana. A new look at the scientific evidence. Oxford University Press; New York: 2002.

NIH-PA Author Manuscript NIH-PA Author Manuscript100. NIH-PA Author Manuscript Aggarwal SK, Carter GT, Sullivan MD, ZumBrunnen C, Morrill R, Mayer JD. Medicinal use of cannabis in the United States: historical perspectives, current trends, and future directions. Journal of opioid management. 2009; 5(3):153–68. [PubMed: 19662925] 101. Fankhauser, M. History of cannabis in Western Medicine. Cannabis and Cannabinoids. The Haworth Integrative Healing Press; New York: 2002. p. 37-51. 102. Hanus LO. Pharmacological and therapeutic secrets of plant and brain (endo)cannabinoids. Medicinal research reviews. 2009; 29(2):213–71. [PubMed: 18777572] 103. Escohotado, A. Historia general de las drogas. Espasa Calpe; Madrid: 1998. 104. Reilly D, Didcott P, Swift W, Hall W. Long-term cannabis use: characteristics of users in an Australian rural area. Addiction. 1998; 93(6):837–46. [PubMed: 9744119] 105. Boys A, Marsden J, Griffiths P, Fountain J, Stillwell G, Strang J. Substance use among young people: the relationship between perceived functions and intentions. Addiction. 1999; 94(7): 1043–50. [PubMed: 10707442] 106. Latimer W, Zur J. Epidemiologic trends of adolescent use of alcohol, tobacco, and other drugs. Child and adolescent psychiatric clinics of North America. 2010; 19(3):451–64. [PubMed: 20682214] 107. Rinaldi-Carmona M, Barth F, Heaulme M, Shire D, Calandra B, Congy C, et al. SR141716A, a potent and selective antagonist of the brain cannabinoid receptor. FEBS Lett. 1994; 350(2-3): 240–4. [PubMed: 8070571] 108. Kendler KS, Prescott CA. Cannabis use, abuse, and dependence in a population-based sample of female twins. The American journal of psychiatry. 1998; 155(8):1016–22. [PubMed: 9699687] 109. Swift W, Hall W, Teesson M. Cannabis use and dependence among Australian adults: results from the National Survey of Mental Health and Wellbeing. Addiction. 2001; 96(5):737–48. [PubMed: 11331032] 110. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 4th ed.. American Psychiatric Association Press; Washington: 2000. 111. Nutt DJ, Ballenger JC, Sheehan D, Wittchen HU. Generalized anxiety disorder: comorbidity, comparative biology and treatment. Int J Neuropsychopharmacol. 2002; 5(4):315–25. [PubMed: 12466031] 112. Hollister LE. Health aspects of cannabis. Pharmacol Rev. 1986; 38(1):1–20. [PubMed: 3520605] 113. Hall W, Solowij N. Adverse effects of cannabis. Lancet. 1998; 352(9140):1611–6. [PubMed: 9843121] 114. Patton GC, Coffey C, Carlin JB, Degenhardt L, Lynskey M, Hall W. Cannabis use and mental health in young people: cohort study. BMJ. 2002; 325(7374):1195–8. [PubMed: 12446533] 115. Green B, Kavanagh D, Young R. Being stoned: a review of self-reported cannabis effects. Drug Alcohol Rev. 2003; 22(4):453–60. [PubMed: 14660135] 116. Dannon PN, Lowengrub K, Amiaz R, Grunhaus L, Kotler M. Comorbid cannabis use and panic disorder: short term and long term follow-up study. Hum Psychopharmacol. 2004; 19(2):97–101. [PubMed: 14994319] 117. Gruber AJ, Pope HG Jr, Brown ME. Do patients use marijuana as an antidepressant? Depression. 1996; 4(2):77–80. [PubMed: 9160645] 118. Stewart SH, Karp J, Pihl RO, Peterson RA. Anxiety sensitivity and self-reported reasons for drug use. J Subst Abuse. 1997; 9:223–40. [PubMed: 9494951] 119. Grinspoon L, Bakalar JB. The use of cannabis as a mood stabilizer in bipolar disorder: anecdotal evidence and the need for clinical research. J Psychoactive Drugs. 1998; 30(2):171–7. [PubMed: 9692379] 120. Ogborne AC, Smart RG, Adlaf EM. Self-reported medical use of marijuana: a survey of the general population. CMAJ. 2000; 162(12):1685–6. [PubMed: 10870496] 121. Agosti V, Nunes E, Levin F. Rates of psychiatric comorbidity among U. Am J Drug Alcohol Abuse. 2002; 28(4):643–52. [PubMed: 12492261]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 18

122. Ashton CH, Moore PB, Gallagher P, Young AH. Cannabinoids in bipolar affective disorder: a review and discussion of their therapeutic potential. J Psychopharmacol. 2005; 19(3):293–300. [PubMed: 15888515] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 123. Arendt M, Rosenberg R, Fjordback L, Brandholdt J, Foldager L, Sher L, et al. Testing the self- medication hypothesis of depression and aggression in cannabis-dependent subjects. Psychol Med. 2007; 37(7):935–45. [PubMed: 17202003] 124. Tournier M, Sorbara F, Gindre C, Swendsen JD, Verdoux H. Cannabis use and anxiety in daily life: a naturalistic investigation in a non-clinical population. Psychiatry Res. 2003; 118(1):1–8. [PubMed: 12759155] 125. Wittchen HU, Frohlich C, Behrendt S, Gunther A, Rehm J, Zimmermann P, et al. Cannabis use and cannabis use disorders and their relationship to mental disorders: a 10-year prospective- longitudinal community study in adolescents. Drug Alcohol Depend. 2007; 88(Suppl 1):S60–70. [PubMed: 17257779] 126. Bonn-Miller MO, Vujanovic AA, Feldner MT, Bernstein A, Zvolensky MJ. Posttraumatic stress symptom severity predicts marijuana use coping motives among traumatic event-exposed marijuana users. J Trauma Stress. 2007; 20(4):577–86. [PubMed: 17721963] 127. Buckner JD, Schmidt NB, Lang AR, Small JW, Schlauch RC, Lewinsohn PM. Specificity of social anxiety disorder as a risk factor for alcohol and cannabis dependence. J Psychiatr Res. 2008; 42(3):230–9. [PubMed: 17320907] 128. Bremner JD, Southwick SM, Darnell A, Charney DS. Chronic PTSD in Vietnam combat veterans: course of illness and substance abuse. Am J Psychiatry. 1996; 153(3):369–75. [PubMed: 8610824] 129. Jacobsen LK, Southwick SM, Kosten TR. Substance use disorders in patients with posttraumatic stress disorder: a review of the literature. Am J Psychiatry. 2001; 158(8):1184–90. [PubMed: 11481147] 130. Reynolds M, Mezey G, Chapman M, Wheeler M, Drummond C, Baldacchino A. Co-morbid post- traumatic stress disorder in a substance misusing clinical population. Drug Alcohol Depend. 2005; 77(3):251–8. [PubMed: 15734225] 131. Cougle JR, Bonn-Miller MO, Vujanovic AA, Zvolensky MJ, Hawkins KA. Posttraumatic stress disorder and Cannabis use in a nationally representative sample. Psychol Addict Behav. 2011 132. Fontenelle LF, Hasler G. The analytical epidemiology of obsessive-compulsive disorder: risk factors and correlates. Prog Neuropsychopharmacol Biol Psychiatry. 2008; 32(1):1–15. [PubMed: 17689849] 133. Schindler F, Anghelescu I, Regen F, Jockers-Scherubl M. Improvement in refractory obsessive compulsive disorder with dronabinol. Am J Psychiatry. 2008; 165(4):536–7. [PubMed: 18381920] 134. Grant JE, Odlaug BL, Chamberlain SR, Kim SW. Dronabinol, a cannabinoid agonist, reduces hair pulling in trichotillomania: a pilot study. Psychopharmacology. 2011 135. Zvolensky MJ, Lewinsohn P, Bernstein A, Schmidt NB, Buckner JD, Seeley J, et al. Prospective associations between cannabis use, abuse, and dependence and panic attacks and disorder. J Psychiatr Res. 2008; 42(12):1017–23. [PubMed: 18076905] 136. Strakowski SM, DelBello MP, Fleck DE, Arndt S. The impact of substance abuse on the course of bipolar disorder. Biological psychiatry. 2000; 48(6):477–85. [PubMed: 11018221] 137. Baethge C, Baldessarini RJ, Khalsa HM, Hennen J, Salvatore P, Tohen M. Substance abuse in first-episode bipolar I disorder: indications for early intervention. Am J Psychiatry. 2005; 162(5): 1008–10. [PubMed: 15863809] 138. Strakowski SM, DelBello MP, Fleck DE, Adler CM, Anthenelli RM, Keck PE Jr. et al. Effects of co-occurring cannabis use disorders on the course of bipolar disorder after a first hospitalization for mania. Arch Gen Psychiatry. 2007; 64(1):57–64. [PubMed: 17199055] 139. Bortolato M, Bini V, Tambaro S. Vulnerability Factors for the Psychiatric and Behavioral Effects of Cannabis. Pharmaceuticals. 2010; 3(9):2799–820. 140. Thomas H. A community survey of adverse effects of cannabis use. Drug Alcohol Depend. 1996; 42(3):201–7. [PubMed: 8912803]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 19

141. Atha, MJ.; Blanchard, S. Regular Users - Self-reported drug consumption patterns and attitudes to drugs among 1333 regular cannabis users. Independent Drug Monitoring Unit; UK: 1997.

NIH-PA Author Manuscript NIH-PA Author Manuscript142. NIH-PA Author Manuscript D’Souza DC, Perry E, MacDougall L, Ammerman Y, Cooper T, Wu YT, et al. The psychotomimetic effects of intravenous delta-9-tetrahydrocannabinol in healthy individuals: implications for psychosis. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2004; 29(8):1558–72. [PubMed: 15173844] 143. Favrat B, Menetrey A, Augsburger M, Rothuizen LE, Appenzeller M, Buclin T, et al. Two cases of “cannabis acute psychosis” following the administration of oral cannabis. BMC Psychiatry. 2005; 5:17. [PubMed: 15804348] 144. Glass RM, Uhlenhuth EH, Hartel FW, Schuster CR, Fischman MW. A single dose study of nabilone, a synthetic cannabinoid. Psychopharmacology. 1980; 71(2):137–42. [PubMed: 6108592] 145. Fabre LF, McLendon D. The efficacy and safety of nabilone (a synthetic cannabinoid) in the treatment of anxiety. J Clin Pharmacol. 1981; 21(8-9 Suppl):377S–82S. [PubMed: 6117575] 146. Schifano F, Corozza O, Deluca P, Davey Z, Di Furia L, Farre M, et al. Psychoactive drug or mystical incense? Overview of the online available information on Spice products. International Journal of Culture and Mental Health. 2009; 2(2):137–44. 147. Auwarter V, Dresen S, Weinmann W, Muller M, Putz M, Ferreiros N. ‘Spice’ and other herbal blends: harmless incense or cannabinoid designer drugs? J Mass Spectrom. 2009; 44(5):832–7. [PubMed: 19189348] 148. Bebarta SV, Varney S, Sessions D, Barry D, Borys D. Spice: A New “Legal” Herbal Mixture Abused by Young Active Duty Military Personnel. Clinical Toxicology. 2010; 48(6):632–3. 149. Muller H, Sperling W, Kohrmann M, Huttner HB, Kornhuber J, Maler JM. The synthetic cannabinoid Spice as a trigger for an acute exacerbation of cannabis induced recurrent psychotic episodes. Schizophr Res. 2010; 118(1-3):309–10. [PubMed: 20056392] 150. Schneir AB, Cullen J, Ly BT. “Spice” girls: synthetic cannabinoid intoxication. J Emerg Med. 2011; 40(3):296–9. [PubMed: 21167669] 151. Benford DM, Caplan JP. Psychiatric sequelae of spice, K2, and synthetic cannabinoid receptor agonists. Psychosomatics. 2011; 52(3):295. [PubMed: 21565605] 152. Braida D, Limonta V, Malabarba L, Zani A, Sala M. 5-HT1A receptors are involved in the anxiolytic effect of Delta9-tetrahydrocannabinol and AM 404, the anandamide transport inhibitor, in Sprague-Dawley rats. Eur J Pharmacol. 2007; 555(2-3):156–63. [PubMed: 17116299] 153. Valjent E, Mitchell JM, Besson MJ, Caboche J, Maldonado R. Behavioural and biochemical evidence for interactions between Delta 9-tetrahydrocannabinol and nicotine. Br J Pharmacol. 2002; 135(2):564–78. [PubMed: 11815392] 154. Berrendero F, Maldonado R. Involvement of the opioid system in the anxiolytic-like effects induced by Delta(9)-tetrahydrocannabinol. Psychopharmacology. 2002; 163(1):111–7. [PubMed: 12185408] 155. Patel S, Hillard CJ. Pharmacological evaluation of cannabinoid receptor ligands in a mouse model of anxiety: further evidence for an anxiolytic role for endogenous cannabinoid signaling. J Pharmacol Exp Ther. 2006; 318(1):304–11. [PubMed: 16569753] 156. Haller J, Varga B, Ledent C, Freund TF. CB1 cannabinoid receptors mediate anxiolytic effects: convergent genetic and pharmacological evidence with CB1-specific agents. Behavioural pharmacology. 2004; 15(4):299–304. [PubMed: 15252281] 157. Onaivi ES, Green MR, Martin BR. Pharmacological characterization of cannabinoids in the elevated plus maze. J Pharmacol Exp Ther. 1990; 253(3):1002–9. [PubMed: 2162942] 158. McGregor IS, Issakidis CN, Prior G. Aversive effects of the synthetic cannabinoid CP 55,940 in rats. Pharmacol Biochem Behav. 1996; 53(3):657–64. [PubMed: 8866969] 159. Rodriguez de Fonseca F, Rubio P, Menzaghi F, Merlo-Pich E, Rivier J, Koob GF, et al. Corticotropin-releasing factor (CRF) antagonist [D-Phe12,Nle21,38,C alpha MeLeu37]CRF attenuates the acute actions of the highly potent cannabinoid receptor agonist HU-210 on defensive-withdrawal behavior in rats. J Pharmacol Exp Ther. 1996; 276(1):56–64. [PubMed: 8558457]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 20

160. Marco EM, Perez-Alvarez L, Borcel E, Rubio M, Guaza C, Ambrosio E, et al. Involvement of 5- HT1A receptors in behavioural effects of the cannabinoid receptor agonist CP 55,940 in male rats. Behavioural pharmacology. 2004; 15(1):21–7. [PubMed: 15075623] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 161. Celerier E, Ahdepil T, Wikander H, Berrendero F, Nyberg F, Maldonado R. Influence of the anabolic-androgenic steroid nandrolone on cannabinoid dependence. Neuropharmacology. 2006; 50(7):788–806. [PubMed: 16443242] 162. Genn RF, Tucci S, Marco EM, Viveros MP, File SE. Unconditioned and conditioned anxiogenic effects of the cannabinoid receptor agonist CP 55,940 in the social interaction test. Pharmacol Biochem Behav. 2004; 77(3):567–73. [PubMed: 15006468] 163. Bortolato M, Piomelli D. Chapter 4. 5 The endocannabinoid system and anxiety responses. Handbook of Behavioral Neuroscience. Elsevier. 2008:303–324. 164. Crippa JA, Zuardi AW, Martin-Santos R, Bhattacharyya S, Atakan Z, McGuire P, et al. Cannabis and anxiety: a critical review of the evidence. Hum Psychopharmacol. 2009; 24(7):515–23. [PubMed: 19693792] 165. Katona I, Sperlagh B, Sik A, Kafalvi A, Vizi ES, Mackie K, et al. Presynaptically located CB1 cannabinoid receptors regulate GABA release from axon terminals of specific hippocampal interneurons. The Journal of neuroscience: the official journal of the Society for Neuroscience. 1999; 19(11):4544–58. [PubMed: 10341254] 166. Azad SC, Monory K, Marsicano G, Cravatt BF, Lutz B, Zieglgansberger W, et al. Circuitry for associative plasticity in the amygdala involves endocannabinoid signaling. The Journal of neuroscience: the official journal of the Society for Neuroscience. 2004; 24(44):9953–61. [PubMed: 15525780] 167. Moreira FA, Aguiar DC, Guimaraes FS. Anxiolytic-like effect of cannabinoids injected into the rat dorsolateral periaqueductal gray. Neuropharmacology. 2007; 52(3):958–65. [PubMed: 17156799] 168. Allen, JRA.; Amegadzie, AK.; Gardinier, KM.; Gregory, GS.; Hitchcock, SA.; Hoogestraat, SA., et al. CB1 Modulator compounds. 2008. US7595339 169. Josephus, LHM.; Henderik, WC.; Cornelis, GK. Tetrasubstituted imidazole derivatives as cannabinoid CB1 receptor modulators with a high CB1/CB2 receptor subtype selectivity. 2009. US7524867 170. Despres JP, Ross R, Boka G, Almeras N, Lemieux I, ADAGIO-Lipids Investigator. Effect of Rimonabant on the High-Triglyceride/Low-HDL-Cholesterol Dyslipidemia, Intraabdominal Adiposity, and Liver Fat. Arterioscler Thromb Vasc Biol. 2009; 29(3):416–23. [PubMed: 19112166] 171. Cahill K, Ussher M. Cannabinoid type 1 receptor antagonists (rimonabant) for smoking cessation. Cochrane Database Syst Rev. 2007; 3:CD005353. [PubMed: 17636794] 172. Navarro M, Hernandez E, Munoz RM, del Arco I, Villanua MA, Carrera MR, et al. Acute administration of the CB1 cannabinoid receptor antagonist SR 141716A induces anxiety-like responses in the rat. Neuroreport. 1997; 8(2):491–6. [PubMed: 9080435] 173. Arevalo C, de Miguel R, Hernandez-Tristan R. Cannabinoid effects on anxiety-related behaviours and hypothalamic neurotransmitters. Pharmacol Biochem Behav. 2001; 70(1):123–31. [PubMed: 11566149] 174. Patel S, Cravatt BF, Hillard CJ. Synergistic interactions between cannabinoids and environmental stress in the activation of the central amygdala. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2005; 30(3):497–507. [PubMed: 15280883] 175. Bortolato M, Campolongo P, Mangieri RA, Scattoni ML, Frau R, Trezza V, et al. Anxiolytic-like properties of the anandamide transport inhibitor AM404. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2006; 31(12):2652–9. [PubMed: 16541083] 176. Van Gaal LF, Rissanen AM, Scheen AJ, Ziegler O, Rossner S. Effects of the cannabinoid-1 receptor blocker rimonabant on weight reduction and cardiovascular risk factors in overweight patients: 1-year experience from the RIO-Europe study. Lancet. 2005; 365(9468):1389–97. [PubMed: 15836887]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 21

177. Christensen R, Kristensen PK, Bartels EM, Bliddal H, Astrup A. Efficacy and safety of the weight-loss drug rimonabant: a meta-analysis of randomised trials. Lancet. 2007; 370(9600): 1706–13. [PubMed: 18022033] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 178. Rucker D, Padwal R, Li SK, Curioni C, Lau DC. Long term pharmacotherapy for obesity and overweight: updated meta-analysis. BMJ. 2007; 335(7631):1194–9. [PubMed: 18006966] 179. Van Gaal LF, Scheen AJ, Rissanen AM, Rossner S, Hanotin C, Ziegler O. Long-term effect of CB1 blockade with rimonabant on cardiometabolic risk factors: two year results from the RIO- Europe Study. Eur Heart J. 2008; 29(14):1761–71. [PubMed: 18417461] 180. Pertwee RG. Inverse agonism and neutral antagonism at cannabinoid CB1 receptors. Life sciences. 2005; 76(12):1307–24. [PubMed: 15670612] 181. Greig, IR.; Ross, RA.; Pertwee, RG. 1,5-diaryl-pyrazoles as cannabinoid receptor neutral antagonists useful as therapeutic agents. 2010. US0022611 182. Sink KS, Segovia KN, Collins LE, Markus EJ, Vemuri VK, Makriyannis A, et al. The CB1 inverse agonist AM251, but not the CB1 antagonist AM4113, enhances retention of contextual fear conditioning in rats. Pharmacol Biochem Behav. 2010; 95(4):479–84. [PubMed: 20347865] 183. Benito C, Nunez E, Tolon RM, Carrier EJ, Rabano A, Hillard CJ, et al. Cannabinoid CB2 receptors and fatty acid amide hydrolase are selectively overexpressed in neuritic plaque- associated glia in Alzheimer’s disease brains. J Neurosci. 2003; 23(35):11136–41. [PubMed: 14657172] 184. Benito C, Romero JP, Tolon RM, Clemente D, Docagne F, Hillard CJ, et al. Cannabinoid CB1 and CB2 receptors and fatty acid amide hydrolase are specific markers of plaque cell subtypes in human multiple sclerosis. J Neurosci. 2007; 27(9):2396–402. [PubMed: 17329437] 185. Yiangou Y, Facer P, Durrenberger P, Chessell IP, Naylor A, Bountra C, et al. COX-2, CB2 and P2X7-immunoreactivities are increased in activated microglial cells/macrophages of multiple sclerosis and amyotrophic lateral sclerosis spinal cord. BMC Neurol. 2006; 6:12. [PubMed: 16512913] 186. Onaivi ES, Ishiguro H, Gong JP, Patel S, Meozzi PA, Myers L, et al. Brain neuronal CB2 cannabinoid receptors in drug abuse and depression: from mice to human subjects. PLoS One. 2008; 3(2):e1640. [PubMed: 18286196] 187. Garcia-Gutierrez MS, Manzanares J. Overexpression of CB2 cannabinoid receptors decreased vulnerability to anxiety and impaired anxiolytic action of alprazolam in mice. J Psychopharmacol. 2011; 25(1):111–20. [PubMed: 20837564] 188. Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, et al. Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2010; 35(3):764–74. [PubMed: 19924114] 189. Zuardi AW, Finkelfarb E, Bueno OF, Musty RE, Karniol IG. Characteristics of the stimulus produced by the mixture of cannabidiol with delta 9-tetrahydrocannabinol. Archives internationales de pharmacodynamie et de therapie. 1981; 249(1):137–46. [PubMed: 6261703] 190. Vann RE, Gamage TF, Warner JA, Marshall EM, Taylor NL, Martin BR, et al. Divergent effects of cannabidiol on the discriminative stimulus and place conditioning effects of Delta(9)- tetrahydrocannabinol. Drug and alcohol dependence. 2008; 94(1-3):191–8. [PubMed: 18206320] 191. Malone DT, Jongejan D, Taylor DA. Cannabidiol reverses the reduction in social interaction produced by low dose Delta(9)-tetrahydrocannabinol in rats. Pharmacology, biochemistry, and behavior. 2009; 93(2):91–6. 192. Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG. Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology. 1982; 76(3): 245–50. [PubMed: 6285406] 193. Reid MJ, Bornheim LM. Cannabinoid-induced alterations in brain disposition of drugs of abuse. Biochemical pharmacology. 2001; 61(11):1357–67. [PubMed: 11331071] 194. Hayakawa K, Mishima K, Hazekawa M, Sano K, Irie K, Orito K, et al. Cannabidiol potentiates pharmacological effects of Delta(9)-tetrahydrocannabinol via CB(1) receptor-dependent mechanism. Brain research. 2008; 1188:157–64. [PubMed: 18021759]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 22

195. Klein C, Karanges E, Spiro A, Wong A, Spencer J, Huynh T, et al. Cannabidiol potentiates Delta(9)-tetrahydrocannabinol (THC) behavioural effects and alters THC pharmacokinetics during acute and chronic treatment in adolescent rats. Psychopharmacology. 2011 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 196. Bornheim LM, Correia MA. Selective inactivation of mouse liver cytochrome P-450IIIA by cannabidiol. Molecular pharmacology. 1990; 38(3):319–26. [PubMed: 2402224] 197. Bornheim LM, Kim KY, Li J, Perotti BY, Benet LZ. Effect of cannabidiol pretreatment on the kinetics of tetrahydrocannabinol metabolites in mouse brain. Drug metabolism and disposition: the biological fate of chemicals. 1995; 23(8):825–31. [PubMed: 7493549] 198. Zuardi AW, Karniol IG. Effects on variable-interval performance in rats of delta 9- tetrahydrocannabinol and cannabidiol, separately and in combination. Braz J Med Biol Res. 1983; 16(2):141–6. [PubMed: 6317104] 199. Musty, RE.; Conti, LH.; Mechoulam, R. Anxiolytic properties of cannabidiol. In: Harvey, DJ., editor. Marihuana ‘84. Proceedings of the Oxford Symposium on Cannabis. IRL Press Limited; Oxford, UK: 1984. p. 713-719. 200. Guimaraes FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety effect of cannabidiol in the elevated plus-maze. Psychopharmacology. 1990; 100(4):558–9. [PubMed: 1969666] 201. Casarotto PC, Gomes FV, Resstel LB, Guimaraes FS. Cannabidiol inhibitory effect on marble- burying behaviour: involvement of CB1 receptors. Behavioural pharmacology. 2010; 21(4):353– 8. [PubMed: 20695034] 202. Bergamaschi MM, Queiroz RH, Chagas MH, de Oliveira DC, De Martinis BS, Kapczinski F, et al. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2011; 36(6):1219–26. [PubMed: 21307846] 203. Crippa JA, Derenusson GN, Ferrari TB, Wichert-Ana L, Duran FL, Martin-Santos R, et al. Neural basis of anxiolytic effects of cannabidiol (CBD) in generalized social anxiety disorder: a preliminary report. J Psychopharmacol. 2011; 25(1):121–30. [PubMed: 20829306] 204. Moreira FA, Aguiar DC, Guimaraes FS. Anxiolytic-like effect of cannabidiol in the rat Vogel conflict test. Prog Neuropsychopharmacol Biol Psychiatry. 2006; 30(8):1466–71. [PubMed: 16876926] 205. Silveira Filho NG, Tufik S. Comparative effects between cannabidiol and diazepam on neophobia, food intake and conflict behavior. Research Communications in Psychology, Psychiatry and Behavior. 1981; 6:25–6. 206. Gomes FV, Casarotto PC, Resstel LB, Guimaraes FS. Facilitation of CB1 receptor-mediated neurotransmission decreases marble burying behavior in mice. Prog Neuropsychopharmacol Biol Psychiatry. 2011; 35(2):434–8. [PubMed: 21111767] 207. Fusar-Poli P, Crippa JA, Bhattacharyya S, Borgwardt SJ, Allen P, Martin-Santos R, et al. Distinct effects of {delta}9-tetrahydrocannabinol and cannabidiol on neural activation during emotional processing. Arch Gen Psychiatry. 2009; 66(1):95–105. [PubMed: 19124693] 208. Campos AC, Guimaraes FS. Involvement of 5HT1A receptors in the anxiolytic-like effects of cannabidiol injected into the dorsolateral periaqueductal gray of rats. Psychopharmacology. 2008; 199(2):223–30. [PubMed: 18446323] 209. de Paula Soares V, Campos AC, Bortoli VC, Zangrossi H Jr. Guimaraes FS, Zuardi AW. Intra- dorsal periaqueductal gray administration of cannabidiol blocks panic-like response by activating 5-HT1A receptors. Behavioural brain research. 2010; 213(2):225–9. [PubMed: 20457188] 210. Bortolato M, Mangieri RA, Fu J, Kim JH, Arguello O, Duranti A, et al. Antidepressant-like activity of the fatty acid amide hydrolase inhibitor URB597 in a rat model of chronic mild stress. Biological psychiatry. 2007; 62(10):1103–10. [PubMed: 17511970] 211. Glaser ST, Abumrad NA, Fatade F, Kaczocha M, Studholme KM, Deutsch DG. Evidence against the presence of an anandamide transporter. Proceedings of the National Academy of Sciences of the United States of America. 2003; 100(7):4269–74. [PubMed: 12655057] 212. Moore SA, Nomikos GG, Dickason-Chesterfield AK, Schober DA, Schaus JM, Ying BP, et al. Identification of a high-affinity binding site involved in the transport of endocannabinoids. Proceedings of the National Academy of Sciences of the United States of America. 2005; 102(49):17852–7. [PubMed: 16314570]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 23

213. Davis, MR.; Ressler, KJ.; Chhatwal, JP.; McDevitt, JP. Augmentation of psychotherapy with cannabinoid reuptake inhibitors. 2006. US0084659

NIH-PA Author Manuscript NIH-PA Author Manuscript214. NIH-PA Author Manuscript Panlilio LV, Justinova Z, Goldberg SR. Animal models of cannabinoid reward. Br J Pharmacol. 2010; 160(3):499–510. [PubMed: 20590560] 215. Kathuria S, Gaetani S, Fegley D, Valino F, Duranti A, Tontini A, et al. Modulation of anxiety through blockade of anandamide hydrolysis. Nat Med. 2003; 9(1):76–81. [PubMed: 12461523] 216. Patel S, Roelke CT, Rademacher DJ, Cullinan WE, Hillard CJ. Endocannabinoid signaling negatively modulates stress-induced activation of the hypothalamic-pituitary-adrenal axis. Endocrinology. 2004; 145(12):5431–8. [PubMed: 15331569] 217. Gaetani S, Dipasquale P, Romano A, Righetti L, Cassano T, Piomelli D, et al. The endocannabinoid system as a target for novel anxiolytic and antidepressant drugs. Int Rev Neurobiol. 2009; 85:57–72. [PubMed: 19607961] 218. Naidu PS, Booker L, Cravatt BF, Lichtman AH. Synergy between enzyme inhibitors of fatty acid amide hydrolase and cyclooxygenase in visceral nociception. J Pharmacol Exp Ther. 2009; 329(1):48–56. [PubMed: 19118134] 219. Lisboa SF, Resstel LB, Aguiar DC, Guimaraes FS. Activation of cannabinoid CB1 receptors in the dorsolateral periaqueductal gray induces anxiolytic effects in rats submitted to the Vogel conflict test. Eur J Pharmacol. 2008; 593(1-3):73–8. [PubMed: 18691568] 220. Rubino T, Realini N, Castiglioni C, Guidali C, Vigano D, Marras E, et al. Role in anxiety behavior of the endocannabinoid system in the prefrontal cortex. Cereb Cortex. 2008; 18(6): 1292–301. [PubMed: 17921459] 221. Haller J, Barna I, Barsvari B, Gyimesi Pelczer K, Yasar S, Panlilio LV, et al. Interactions between environmental aversiveness and the anxiolytic effects of enhanced cannabinoid signaling by FAAH inhibition in rats. Psychopharmacology. 2009; 204(4):607–16. [PubMed: 19259645] 222. Justinova Z, Mangieri RA, Bortolato M, Chefer SI, Mukhin AG, Clapper JR, et al. Fatty acid amide hydrolase inhibition heightens anandamide signaling without producing reinforcing effects in primates. Biological psychiatry. 2008; 64(11):930–7. [PubMed: 18814866] 223. Vandevoorde S. Overview of the chemical families of fatty acid amide hydrolase and monoacylglycerol lipase inhibitors. Curr Top Med Chem. 2008; 8(3):247–67. [PubMed: 18289091] 224. Clapper JR, Vacondio F, King AR, Duranti A, Tontini A, Silva C, et al. A second generation of carbamate-based fatty acid amide hydrolase inhibitors with improved activity in vivo. ChemMedChem. 2009; 4(9):1505–13. [PubMed: 19637155] 225. Sing-Yuen, SK.; Ka, X.; Hongfeng, D. (Oxime)carbamoyl fatty acid amide hydrolase inhibitors. 2005. US6949574 226. Ahn K, Johnson DS, Fitzgerald LR, Liimatta M, Arendse A, Stevenson T, et al. Novel mechanistic class of fatty acid amide hydrolase inhibitors with remarkable selectivity. Biochemistry. 2007; 46(45):13019–30. [PubMed: 17949010] 227. Seillier A, Advani T, Cassano T, Hensler JG, Giuffrida A. Inhibition of fatty-acid amide hydrolase and CB1 receptor antagonism differentially affect behavioural responses in normal and PCP-treated rats. Int J Neuropsychopharmacol. 2010; 13(3):373–86. [PubMed: 19607756] 228. Sokolic L, Long LE, Hunt GE, Arnold JC, McGregor IS. Disruptive effects of the prototypical cannabinoid Delta-tetrahydrocannabinol and the fatty acid amide inhibitor URB-597 on go/no-go auditory discrimination performance and olfactory reversal learning in rats. Behavioural pharmacology. 2011; 22(3):191–202. [PubMed: 21512341] 229. Sciolino NR, Zhou W, Hohmann AG. Enhancement of endocannabinoid signaling with JZL184, an inhibitor of the 2-arachidonoylglycerol hydrolyzing enzyme monoacylglycerol lipase, produces anxiolytic effects under conditions of high environmental aversiveness in rats. Pharmacol Res. 2011; 64(3):226–34. [PubMed: 21600985] 230. Kinsey SG, O’Neal ST, Long JZ, Cravatt BF, Lichtman AH. Inhibition of endocannabinoid catabolic enzymes elicits anxiolytic-like effects in the marble burying assay. Pharmacol Biochem Behav. 2011; 98(1):21–7. [PubMed: 21145341]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 24

231. Sutt S, Raud S, Areda T, Reimets A, Koks S, Vasar E. Cat odour-induced anxiety--a study of the involvement of the endocannabinoid system. Psychopharmacology. 2008; 198(4):509–20. [PubMed: 17882402] NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript 232. Suarez J, Rivera P, Llorente R, Romero-Zerbo SY, Bermudez-Silva FJ, de Fonseca FR, et al. Early maternal deprivation induces changes on the expression of 2-AG biosynthesis and degradation enzymes in neonatal rat hippocampus. Brain research. 2010; 1349:162–73. [PubMed: 20599824] 233. Olmstead, MC.; Paquette, JJ. Methods and therapies for potentiating therapeutic activities of a cannabinoid receptor agonist via administration of a cannabinoid receptor antagonist. 2007. US0060638 234. Aragona M, Onesti E, Tomassini V, Conte A, Gupta S, Gilio F, et al. Psychopathological and cognitive effects of therapeutic cannabinoids in multiple sclerosis: a double-blind, placebo controlled, crossover study. Clin Neuropharmacol. 2009; 32(1):41–7. [PubMed: 18978501] 235. Karschner EL, Darwin WD, McMahon RP, Liu F, Wright S, Goodwin RS, et al. Subjective and physiological effects after controlled Sativex and oral THC administration. Clin Pharmacol Ther. 2011; 89(3):400–7. [PubMed: 21289620] 236. Long JZ, Nomura DK, Vann RE, Walentiny DM, Booker L, Jin X, et al. Dual blockade of FAAH and MAGL identifies behavioral processes regulated by endocannabinoid crosstalk in vivo. Proc Natl Acad Sci U S A. 2009; 106(48):20270–5. [PubMed: 19918051] 237. Justinova Z, Yasar S, Redhi GH, Goldberg SR. The endogenous cannabinoid 2- arachidonoylglycerol is intravenously self-administered by squirrel monkeys. J Neurosci. 2011; 31(19):7043–8. [PubMed: 21562266] 238. Weil AT, Zinberg NE, Nelsen JM. Clinical and psychological effects of marihuana in man. Science. 1968; 162(3859):1234–42. [PubMed: 4880784] 239. Jones RT. Marihuana-induced “high”: influence of expectation, setting and previous drug experience. Pharmacol Rev. 1971; 23(4):359–69. [PubMed: 4943950] 240. Tennant FS Jr. Groesbeck CJ. Psychiatric effects of hashish. Arch Gen Psychiatry. 1972; 27(1): 133–6. [PubMed: 5032722] 241. Millman RB, Sbriglio R. Patterns of use and psychopathology in chronic marijuana users. Psychiatr Clin North Am. 1986; 9(3):533–45. [PubMed: 3022257] 242. Dorard G, Berthoz S, Haviland MG, Phan O, Corcos M, Bungener C. Multimethod alexithymia assessment in adolescents and young adults with a cannabis use disorder. Compr Psychiatry. 2008; 49(6):585–92. [PubMed: 18970907] 243. Jones RT, Benowitz NL, Herning RI. Clinical relevance of cannabis tolerance and dependence. J Clin Pharmacol. 1981; 21(8-9 Suppl):143S–52S. [PubMed: 6271820] 244. Ashton CH. Adverse effects of cannabis and cannabinoids. Br J Anaesth. 1999; 83(4):637–49. [PubMed: 10673884] 245. Oviedo A, Glowa J, Herkenham M. Chronic cannabinoid administration alters cannabinoid receptor binding in rat brain: a quantitative autoradiographic study. Brain research. 1993; 616(1-2):293–302. [PubMed: 8395305] 246. Rodriguez de Fonseca F, Gorriti MA, Fernandez-Ruiz JJ, Palomo T, Ramos JA. Downregulation of rat brain cannabinoid binding sites after chronic delta 9-tetrahydrocannabinol treatment. Pharmacol Biochem Behav. 1994; 47(1):33–40. [PubMed: 8115426] 247. Henquet C, Di Forti M, Morrison P, Kuepper R, Murray RM. Gene-environment interplay between cannabis and psychosis. Schizophrenia bulletin. 2008; 34(6):1111–21. [PubMed: 18723841] 248. Murray RM, Morrison PD, Henquet C, Di Forti M. Cannabis, the mind and society: the hash realities. Nature reviews Neuroscience. 2007; 8(11):885–95. 249. Arseneault L, Cannon M, Witton J, Murray RM. Causal association between cannabis and psychosis: examination of the evidence. The British journal of psychiatry: the journal of mental science. 2004; 184:110–7. [PubMed: 14754822] 250. Houston JE, Murphy J, Adamson G, Stringer M, Shevlin M. Childhood sexual abuse, early cannabis use, and psychosis: testing an interaction model based on the National Comorbidity Survey. Schizophrenia bulletin. 2008; 34(3):580–5. [PubMed: 18024467]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 25

251. Caspi A, Moffitt TE, Cannon M, McClay J, Murray R, Harrington H, et al. Moderation of the effect of adolescent-onset cannabis use on adult psychosis by a functional polymorphism in the catechol-O-methyltransferase gene: longitudinal evidence of a gene X environment interaction. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript Biological psychiatry. 2005; 57(10):1117–27. [PubMed: 15866551] 252. Henquet C, Rosa A, Delespaul P, Papiol S, Fananas L, van Os J, et al. COMT ValMet moderation of cannabis-induced psychosis: a momentary assessment study of ‘switching on’ hallucinations in the flow of daily life. Acta psychiatrica Scandinavica. 2009; 119(2):156–60. [PubMed: 18808401] 253. Henquet C, Rosa A, Krabbendam L, Papiol S, Fananas L, Drukker M, et al. An experimental study of catechol-o-methyltransferase Val158Met moderation of delta-9-tetrahydrocannabinol- induced effects on psychosis and cognition. Neuropsychopharmacology: official publication of the American College of Neuropsychopharmacology. 2006; 31(12):2748–57. [PubMed: 16936704] 254. Boucher AA, Arnold JC, Duffy L, Schofield PR, Micheau J, Karl T. Heterozygous neuregulin 1 mice are more sensitive to the behavioural effects of Delta9-tetrahydrocannabinol. Psychopharmacology. 2007; 192(3):325–36. [PubMed: 17333138] 255. Boucher AA, Hunt GE, Micheau J, Huang X, McGregor IS, Karl T, et al. The schizophrenia susceptibility gene neuregulin 1 modulates tolerance to the effects of cannabinoids. Int J Neuropsychopharmacol. 2011; 14(5):631–43. [PubMed: 20701826] 256. Khare M, Taylor AH, Konje JC, Bell SC. Delta9-tetrahydrocannabinol inhibits cytotrophoblast cell proliferation and modulates gene transcription. Molecular human reproduction. 2006; 12(5): 321–33. [PubMed: 16597638]

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 26 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Fig. 1. Chemical structures of the major phytocannabinoids. For more details, see text.

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 27 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Fig. 2. Chemical structures of the synthetic THC analogs CP55,940, CP55,244, CP 47,497 and HU-210. For more details, see text.

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 28 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Fig. 3. Chemical structures of the major endocannabinoids. For more details, see text.

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 29 NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Fig. 4. Chemical structures of endocannabinoid degradation inactivators. For more details, see text.

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 30

Table 1 Current pharmacological strategies for the treatment of anxiety disorders NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

1 Generalized anxiety disorder a. Benzodiazepines b. Buspirone c. Selective serotonin reuptake inhibitors 2 Panic attack a. High-potency benzodiazepines b. Tricyclic antidepressants c. Selective serotonin reuptake inhibitors d. Monoamine oxidase inhibitors 3 Post-traumatic stress disorder a. Selective serotonin reuptake inhibitors b. Low-dose antipsychotic agents 4 Obsessive-compulsive disorder a. Tricyclic antidepressants b. Selective serotonin reuptake inhibitors

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Tambaro and Bortolato Page 31

Table 2 Paradigms for testing of anxiety-like behaviors in rodents NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

1 Unconditioned anxiety a. Tests for social anxiety i. Maternal separation-induced ultrasonic vocalizations (for pups) ii. Social interaction b. Tests based on approach/avoidance conflict i. Novel open field ii. Defensive withdrawal iii. Elevated plus maze iv. Elevated T-maze v. Zero maze vi. Light/dark box vii. Emergence test c. Tests based on antipredator defensive behavior i. Mouse defense test battery ii. Predator urine exposure test iii. Predator exposure test d. Other tests i. Novelty-induced feeding suppression ii. Marble burying iii. Defensive burying 2 Conditioned anxiety a. Tests on conditional fear i. Fear- conditioned freezing ii. Fear-potentiated startle iii. Conditional fear-induced analgesia b. Operant conflict test i. Geiller-Seifter test (conditioned suppression of eating) ii. Vogel test (conditioned suppression of drinking)

Recent Pat CNS Drug Discov. Author manuscript; available in PMC 2013 June 25. Neurotherapeutics (2015) 12:825–836 DOI 10.1007/s13311-015-0387-1

REVIEW

Cannabidiol as a Potential Treatment for Anxiety Disorders

Esther M. Blessing1 & Maria M. Steenkamp1 & Jorge Manzanares1,2 & Charles R. Marmar1

Published online: 4 September 2015 # The American Society for Experimental NeuroTherapeutics, Inc. 2015

Abstract Cannabidiol (CBD), a Cannabis sativa constituent, Introduction is a pharmacologically broad-spectrum drug that in recent years has drawn increasing interest as a treatment for a range Fear and anxiety are adaptive responses essential to coping of neuropsychiatric disorders. The purpose of the current re- with threats to survival. Yet excessive or persistent fear may view is to determine CBD’s potential as a treatment for be maladaptive, leading to disability. Symptoms arising from anxiety-related disorders, by assessing evidence from preclin- excessive fear and anxiety occur in a number of neuropsychi- ical, human experimental, clinical, and epidemiological stud- atric disorders, including generalized anxiety disorder (GAD), ies. We found that existing preclinical evidence strongly sup- panic disorder (PD), post-traumatic stress disorder (PTSD), ports CBD as a treatment for generalized anxiety disorder, social anxiety disorder (SAD), and obsessive–compulsive dis- panic disorder, social anxiety disorder, obsessive–compulsive order (OCD). Notably, PTSD and OCD are no longer classi- disorder, and post-traumatic stress disorder when adminis- fied as anxiety disorders in the recent revision of the Diagnos- tered acutely; however, few studies have investigated chronic tic and Statistical Manual of Mental Disorders-5; however, CBD dosing. Likewise, evidence from human studies sup- excessive anxiety is central to the symptomatology of both ports an anxiolytic role of CBD, but is currently limited to disorders. These anxiety-related disorders are associated with acute dosing, also with few studies in clinical populations. a diminished sense of well-being, elevated rates of unemploy- Overall, current evidence indicates CBD has considerable po- ment and relationship breakdown, and elevated suicide risk tential as a treatment for multiple anxiety disorders, with need [1–3]. Together, they have a lifetime prevalence in the USA for further study of chronic and therapeutic effects in relevant of 29 % [4], the highest of any mental disorder, and constitute clinical populations. an immense social and economic burden [5, 6]. Currently available pharmacological treatments include sero- tonin reuptake inhibitors, serotonin–norepinephrine reuptake in- Keywords Cannabidiol . Endocannabinoids . Anxiety . hibitors, benzodiazepines, monoamine oxidase inhibitors, tricy- Generalized anxiety disorder . Post-traumatic stress disorder clic antidepressant drugs, and partial 5-hydroxytryptamine (5-

HT)1A receptor agonists. Anticonvulsants and atypical antipsy- chotics are also used to treat PTSD. These medications are asso- ciated with limited response rates and residual symptoms, partic- ularly in PTSD, and adverse effects may also limit tolerability and adherence [7–10]. The substantial burden of anxiety-related * Esther M. Blessing disorders and the limitations of current treatments place a high [email protected] priority on developing novel pharmaceutical treatments. Cannabidiol (CBD) is a phytocannabinoid constituent of 9- 1 New York University School of Medicine, New York, NY, USA Cannabis sativa that lacks the psychoactive effects of Δ tet- rahydrocannabinol (THC). CBD has broad therapeutic prop- 2 Instituto de Neurociencias de Alicante, Universidad Miguel Hernández and Consejo Superior de Investigaciones Científicas, erties across a range of neuropsychiatric disorders, stemming Alicante, Spain from diverse central nervous system actions [11, 12]. In recent 826 Blessing et al. years, CBD has attracted increasing interest as a potential additional TRP channels, and glycine receptors [11, 12, 19, anxiolytic treatment [13–15]. The purpose of this review is 21]. In the current review of primary studies, the following to assess evidence from current preclinical, clinical, and epi- receptor-specific actions were found to have been investigated demiological studies pertaining to the potential risks and ben- as potential mediators of CBD’s anxiolytic action: CB1R, efits of CBD as a treatment for anxiety disorders. TRPV1 receptors, and 5-HT1A receptors. Pharmacology rele- vant to these actions is detailed below.

Methods The Endocannabinoid System

A search of MEDLINE (PubMed), PsycINFO, Web of Science Following cloning of the endogenous receptor for THC,

Scopus, and the Cochrane Library databases was conducted for namely the CB1R, endogenous CB1R ligands, or English-language papers published up to 1 January 2015, using Bendocannabinoids^ (eCBs) were discovered, namely anan- the search terms Bcannabidiol^ and Banxiety^ or Bfear^ or damide (AEA) and 2-arachidonoylglycerol (reviewed in [22]).

Bstress^ or Banxiety disorder^ or Bgeneralized anxiety disorder^ The CB1R is an inhibitory Gi/o protein-coupled receptor that is or Bsocial anxiety disorder^ or Bsocial phobia^ or Bpost-trau- mainly localized to nerve terminals, and is expressed on both matic stress disorder^ or Bpanic disorder^ or Bobsessive com- γ-aminobutryic acid-ergic and glutamatergic neurons. eCBs pulsive disorder^. In total, 49 primary preclinical, clinical, or are fatty acid derivatives that are synthesized on demand in epidemiological studies were included. Neuroimaging studies response to neuronal depolarization and Ca2+ influx, via that documented results from anxiety-related tasks, or resting cleavage of membrane phospholipids. The primary mecha- neural activity, were included. Epidemiological or clinical stud- nism by which eCBs regulate synaptic function is retrograde ies that assessed CBD’s effects on anxiety symptoms, or the signaling, wherein eCBs produced by depolarization of the potential protective effects of CBD on anxiety symptoms in- postsynaptic neuron activate presynaptic CB1Rs, leading to duced by cannabis use (where the CBD content of cannabis is inhibition of neurotransmitter release [23]. The BeCB system^ inferred via a higher CBD:THC ratio), were included. includes AEA and 2-arachidonoylglycerol; their respective degradative enzymes fatty acid amide hydroxylase (FAAH)

and monoacylglycerol lipase; the CB1R and related CB2 re- CBD Pharmacology Relevant to Anxiety ceptor (the latter expressed mainly in the periphery); as well as several other receptors activated by eCBs, including the General Pharmacology and Therapeutic Profile TRPV1 receptor, peroxisome proliferator-activated receptor-γ, and G protein-coupled 55 receptor, which func-

Cannabis sativa,aspeciesoftheCannabis genus of flowering tionally interact with CB1R signaling (reviewed in [21, 24]). plants, is one of the most frequently used illicit recreational Interactions with the TRPV1 receptor, in particular, appear to substances in Western culture. The 2 major phyto- cannabinoid be critical in regulating the extent to which eCB release leads constituents with central nervous system activity are THC, re- to inhibition or facilitation of presynaptic neurotransmitter re- sponsible for the euphoric and mind-altering effects, and CBD, lease [25]. The TRPV1 receptor is a postsynaptic cation chan- which lacks these psychoactive effects. Preclinical and clinical nel that underlies sensation of noxious heat in the periphery, studies show CBD possesses a wide range of therapeutic prop- with capsacin (hot chili) as an exogenous ligand. TRPV1 re- erties, including antipsychotic, analgesic, neuroprotective, anti- ceptors are also expressed in the brain, including the amygdala, convulsant, antiemetic, antioxidant, anti-inflammatory, antiar- periaqueductal grey, hippocampus, and other areas [26, 27]. thritic, and antineoplastic properties (see [11, 12, 16–19]for The eCB system regulates diverse physiological functions, reviews). A review of potential side effects in humans found including caloric energy balance and immune function [28]. that CBD was well tolerated across a wide dose range, up to The eCB system is also integral to regulation of emotional 1500 mg/day (orally), with no reported psychomotor slowing, behavior, being essential to forms of synaptic plasticity that negative mood effects, or vital sign abnormalities noted [20]. determine learning and response to emotionally salient, par-

CBD has a broad pharmacological profile, including inter- ticularly highly aversive events [29, 30]. Activation of CB1Rs actions with several receptors known to regulate fear and produces anxiolytic effects in various models of uncondi- anxiety-related behaviors, specifically the cannabinoid type tioned fear, relevant to multiple anxiety disorder symptom

1 receptor (CB1R), the serotonin 5-HT1A receptor, and the domains (reviewed in [30–33]). Regarding conditioned fear, transient receptor potential (TRP) vanilloid type 1 (TRPV1) the effect of CB1R activation is complex: CB1R activation receptor [11, 12, 19, 21]. In addition, CBD may also regulate, may enhance or reduce fear expression, depending on brain directly or indirectly, the peroxisome proliferator-activated locus and the eCB ligand [34]; however, CB1R activation receptor-γ, the orphan G-protein-coupled receptor 55, the e- potently enhances fear extinction [35], and can prevent quilibrative nucleoside transporter, the adenosine transporter, fear reconsolidation. Genetic manipulations that impede Cannabidiol as a Potential Treatment for Anxiety Disorders 827

CB1R activation are anxiogenic [35], and individuals with various other brain areas involved in fear and anxiety eCB system gene polymorphisms that reduce eCB tone— [54, 55]. Mechanisms underlying the anxiolytic effects for example, FAAH gene polymorphisms—exhibit physio- of 5-HT1AR activation are complex, varying between logical, psychological, and neuroimaging features consis- both brain region, and pre- versus postsynaptic locus, tent with impaired fear regulation [36]. Reduction of and are not fully established [56]. While in vitro studies

AEA–CB1R signaling in the amygdala mediates the suggest CBD acts as a direct 5-HT1AR agonist [57], anxiogenic effects of corticotropin-releasing hormone in vivo studies are more consistent with CBD acting

[37], and CB1R activation is essential to negative feedback as an allosteric modulator, or facilitator of 5-HT1A of the neuroendocrine stress response, and protects against signaling [58]. the adverse effects of chronic stress [38, 39]. Finally, chronic stress impairs eCB signaling in the hippocampus and amygdala, leading to anxiety [40, 41], and people Preclinical Evaluations with PTSD show elevated CB1R availability and reduced peripheral AEA, suggestive of reduced eCB tone [42]. Generalized Anxiety Models

Accordingly, CB1R activation has been suggested as a tar- get for anxiolytic drug development [15, 43, 44]. Proposed Relevant studies in animal models are summarized in chro- agents for enhancing CB1R activation include THC, which nological order in Table 1. CBD has been studied in a wide is a potent and direct agonist; synthetic CB1R agonists; FAAH range of animal models of general anxiety, including the inhibitors and other agents that increase eCB availability, as elevated plus maze (EPM), the Vogel-conflict test (VCT), well as nonpsychoactive cannabis phytocannabinoids, includ- and the elevated T maze (ETM). See Table 1 for the anxi- ing CBD. While CBD has low affinity for the CB1R, it func- olytic effect specific to each paradigm. Initial studies of tions as an indirect agonist, potentially via augmentation of CBD in these models showed conflicting results: high

CB1R constitutional activity, or via increasing AEA through (100 mg/kg) doses were ineffective, while low (10 mg/kg) FAAH inhibition (reviewed in [21]). doses were anxiolytic [59, 60]. When tested over a wide Several complexities of the eCB system may impact upon range of doses in further studies, the anxiolytic effects of the potential of CBD and other CB1R-activating agents to serve CBD presented a bell-shaped dose–response curve, with an- as anxiolytic drugs. First, CB1R agonists, including THC and xiolytic effects observed at moderate but not higher doses AEA, have a biphasic effect: low doses are anxiolytic, but [61, 90]. All further studies of acute systemic CBD without higher doses are ineffective or anxiogenic, in both preclinical prior stress showed anxiolytic effects or no effect [62, 65], models in and humans (reviewed in [33, 45]). This biphasic the latter study involving intracerebroventricular rather than profile may stem from the capacity of CB1R agonists to the intraperitoneal route. No anxiogenic effects of acute sys- also activate TRPV1 receptors when administered at a high, temic CBD dosing in models of general anxiety have yet but not low dose, as demonstrated for AEA [46]. Activation been reported. As yet, few studies have examined chronic of TRPV1 receptors is predominantly anxiogenic, and thus a dosing effects of CBD in models of generalized anxiety. critical balance of eCB levels, determining CB1 versus TRPV1 Campos et al. [66] showed that in rat, CBD treatment for activation, is proposed to govern emotional behavior [27, 47]. 21 days attenuated inhibitory avoidance acquisition [83]. CBD acts as a TRPV1 agonist at high concentrations, poten- Long et al. [69] showed that, in mouse, CBD produced tially by interfering with AEA inactivation [48]. In addition to moderate anxiolytic effects in some paradigms, with no ef- dose-dependent activation of TRPV1 channels, the anxiogenic fects in others. versus anxiolytic balance of CB1R agonists also depends on Anxiolytic effects of CBD in models of generalized anxiety dynamic factors, including environmental stressors [33, 49]. have been linked to specific receptor mechanisms and brain regions. The midbrain dorsal periaqueductal gray (DPAG) is

5-HT1A Receptors integral to anxiety, orchestrating autonomic and behavioral responses to threat [91], and DPAG stimulation in humans

The 5-HT1A receptor (5-HT1AR) is an established anxiolytic produces feelings of intense distress and dread [92]. Microin- target. Buspirone and other 5-HT1AR agonists are approved jection of CBD into the DPAG produced anxiolytic effects in for the treatment of GAD, with fair response rates [50]. In the EPM, VGC, and ETM that were partially mediated by preclinical studies, 5-HT1AR agonists are anxiolytic in animal activation of 5-HT1ARs but not by CB1Rs [65, 68]. The bed models of general anxiety [51], prevent the adverse effects of nucleus of the stria terminalis (BNST) serves as a principal stress [52], and enhance fear extinction [53]. Both pre- and output structure of the amygdaloid complex to coordinate postsynaptic 5-HT1ARs are coupled to various members of the sustained fear responses, relevant to anxiety [93]. Anxiolytic Gi/o protein family. They are expressed on serotonergic neurons effects of CBD in the EPM and VCT occurred upon microin- in the raphe, where they exert autoinhibitory function, and jection into the BNST, where they depended on 5-HT1AR 828 Table 1 Preclinical studies

Study Animal Route Dose Model Effect Receptor Involvement

Silveira Filho et al. [59]WR i.p. 100 mg/kg, GSCT No effect NA acute Zuardi et al. [60]WR i.p. 10mg/kg, CER Anxiolytic NA acute Onaivi et al. [61] ICR mice i.p. 0.01, 0.10, 0.50, 1.00, 2.50, 5.00, EPM Anxiolytic Effects ↓ by IP flumazenil, 10.00, 50.00, 100.00 mg/kg, acute unchanged by naloxone Guimaraes et al. [61]WR i.p. 2.5, 5.0, 10.0 and EPM Anxiolytic NA 20.0 mg/kg, acute Moreira et al. [62]WRi.p.2.5,5.0and10.0mg/kg, acute VCT Anxiolytic Effect unchanged by IP flumazenil Resstel et al. [63]WRi.p. 10mg/kg, acute CFC Anxiolytic NA Campos et al. [64] WR dlPAG 15.0, 30.0, 60.0 nmol/0.2 μl, acute EPM Anxiolytic Both effects ↓ by intra-dlPAG VCT Anxiolytic WAY100635 but not intra-dlPAG AM251 Bitencourt et al. [65]WRi.c.v. 2.0 μg/μl CFC Anxiolytic Extinction effect ↓ by 5 min before extinction, acute extinction SR141716A but not EPM before and No effect before CFC capsazepine 24 h after CFC Anxiolytic following CFC Campos et al. [66]WRdlPAG 30, 60 mg/kg, acute EPM Anxiolytic Intra-dlPAG capsazepine renders 60 mg/kg anxiolytic Resstel et al. [67]WRi.p.1,10 or 20 mg/kg, acute RS Anxiolytic, All effects ↓ by systemic ↓ Pressor WAY100635 ↓ Tachycardia EPM 24 h Anxiolytic following RS Soares et al. [68] WR dlPAG 15, 30 or 60nmol, acute ETM Anxiolytic All effects ↓ by intra-dlPAG Panicolytic WAY100635 but not AM251 PAG E-stim Panicolytic Long et al. [69] C57BL/6 J mice i.p. 1, 5, 10, 50mg/kg, chronic, daily/21 d EPM No effect NA L-DT 1 mg/kg anxiolytic SI No effect OF 50 mg/kg anxiolytic Lemos et al. [70]WRi.p. 10mg/kg IP, 30nmol intra-PL and CFC IP and PL anxiolytic IL NA PL intra-IL, acute anxiogenic IL Casarotto et al. [71] C57BL/6 J mice i.p. 15, 30, and 60 mg/kg, MBT Anticompulsive Effect ↓ by IP AM251 but not acute, or subchronic, daily/7 d WAY100635 Gomes et al. [72] WR BNST 15, 30, and 60 nmol, acute EPM Anxiolytic Both effects ↓ by intra BNST VCT Anxiolytic WAY100635

Granjeiro et a l. [73] WR Intracisternal 15, 30, and 60 nmol, acute RS Anxiolytic, ↓Pressor ↓Tachycardia NA al. et Blessing EPM 24 h after RS Anxiolytic Deiana et al. [74]SMi.p. 120mg/kg, acute MBT Anticompulsive NA Oral Uribe-Marino et al. [75]SM i.p. 0.3,3.0, 30.0mg/kg, acute PS Panicolytic NA anbdo saPtnilTetetfrAxeyDisorders Anxiety for Treatment Potential a as Cannabidiol Table 1 (continued)

Study Animal Route Dose Model Effect Receptor Involvement

Stern et al. [76]WR i.p. 3,10, 30 mg/kg Reconsolidation blockade Anxiolytic Effect ↓ by AM251 but not immediately after retrieval, 1and7doldfearmemories WAY100635 acute disrupted Campos et al. [77]WRi.p. 5 mg/kg, subchronic, daily/7 d EPM following PS Anxiolytic Effects ↓ by IP WAY100635 Hsiao et al. [78]WRCeA1 μg/μl REM sleep time ↓ REM sleep suppression NA EPM Anxiolytic OF Anxiolytic Gomes et al. [79]WRBNST15, 30, 60nmol, acute CFC Anxiolytic Both effects ↓ by intra-BNST WAY100635 El Batsh et al. [80] LE-H R i.p. 10mg/kg, chronic, CFC Anxiogenic NA daily/14 d Campos et al. [81] C57BL/6 mice i.p. 30mg/kg2hafterCUS, EPM Anxiolytic Both effects ↓ by AM251 chronic daily/14 d NSF Anxiolytic Do Monte et al. [82] L-E HR IL 1 μgor0.4μg/0.2 μl Extinction of CFC Anxiolytic Effect ↓ by IP rimonabant 5 min before extinction daily/4 d Campos et al. [83]Rati.p. 5mg/kg, chronic, ETM Anxiolytic Panicolytic effect ↓ by daily/21 d Panicolytic intra-dlPAG WAY100635 Almeida et al. [84]Rati.p. 1, 5, 15 mg/kg, acute SI Anxiolytic NA Gomes et al. [85]WRBNST30 and 60 nmol, acute RS Anxiogenic Effect ↓ by WAY100635 ↑ Tachydardia Twardowschy et al. [86]SM i.p. 3mg/kg, acute PS Panicolytic Effects ↓ by IP WAY100635 Focaga et al. [87] WR PL 15, 30, 60 nmol, acute EPM Anxiogenic All effects ↓ by intra PL EPM after RS Anxiolytic WAY100635 CFC Anxiolytic Anxiolytic EPM effect post-RS ↓ by IP metyrapone Nardo et al. [88]SM i.p. 30 mg/kg, acute MBT Anticompulsive NA da Silva et al. [89]WRSNpr 5μg/0.2μl GABAA blockade Panicolytic Both effects ↓ by AM251 in dlSC

Effective doses are in bold

Receptor specific agents: AM251 = cannabinoid receptor type 1 (CB1R) inverse agonist; WAY100635 = 5-hydroxytryptamine 1A antagonist; SR141716A = CB1R antagonist; rimonabant = CB1R antagonist; capsazepine = transient receptor potential vanilloid type 1 antagonist; naloxone = opioid antagonist; flumazenil = GABAA receptor antagonist Anxiolytic effects in models used: CER = reduced fear response; CFC = reduced conditioned freezing; CFC extinction = reduced freezing following extinction training; EPM = reduced % time in open arm; ETM = decreased inhibitory avoidance; L-DT = increased % time in light; VCT = increased licks indicating reduced conflict; NSF = reduced latency to feed; OF = increased % time in center; SI = increased social interaction Anticomplusive effects: MBT = reduced burying

Panicolytic effects: ETM = decreased escape; GABAA blockade in dlSC = defensive immobility, and explosive escape; PAG-E-Stim = increased threshold for escape; PS = reduced explosive escape WR=Wistarrats;SM=Swissmice;L-EHR=Long–Evans hooded rats; i.p. = intraperitoneal; dlPAG = dorsolateral periaqueductal gray; i.c.v. = intracerebroventricular; PL = prelimbic; IL = infralimbic; BNST = bed nucleus of the stria terminalis; CeA = amygdala central nucleus; SNpr = substantia nigra pars reticularis; CUS = chronic unpredictable stress; GSCT = Geller–Seifter conflict test; CER = conditioned emotional response; EPM = elevated plus maze; VCT = Vogel conflict test; CFC = contextual fear conditioning; RS = restraint stress; ETM = elevated T maze; PAG E-stim = electrical stimulation of the dlPAG; L-DT = light–dark test; SI = social interaction; OF = open field; MBT = marble-burying test; PS = predator stress; NSF = novelty suppressed feeding test; GABAA = γ- aminobutyric acid receptor A; dlSC = deep layers superior colliculus; REM = rapid eye movement; NA = not applicable 829 830 Blessing et al. activation [79], and also upon microinjection into the cen- assesses explosive escape and defensive immobility in re- tral nucleus of the amygdala [78]. In the prelimbic cortex, sponse to a boa constrictor snake, also partially via 5-HT1AR which drives expression of fear responses via connections activation; however, more consistent with an anxiogenic ef- with the amygdala [94], CBD had more complex effects: in fect, CBD was also noted to decrease time spent outside the unstressed rats, CBD was anxiogenic in the EPM, partially burrow and increase defensive attention (not shown in via 5-HT1AR receptor activation; however, following acute Table 1)[75, 86] . Finally, CBD, partially via CB1Rs, de- restraint stress, CBD was anxiolytic [87]. Finally, the anxi- creased defensive immobility and explosive escape caused olytic effects of systemic CBD partially depended on by bicuculline-induced neuronal activation in the superior

GABAA receptor activation in the EPM model but not in colliculus [89]. Anticompulsive effects of CBD were investi- the VCT model [61, 62]. gated in marble-burying behavior, conceptualized to model As noted, CBD has been found to have a bell-shaped re- OCD [96]. Acute systemic CBD reduced marble-burying be- sponse curve, with higher doses being ineffective. This may havior for up to 7 days, with no attenuation in effect up to high reflect activation of TRPV1 receptors at higher dose, as block- (120 mg/kg) doses, and effect shown to depend on CB1Rs but ade of TRPV1 receptors in the DPAG rendered a previously not 5-HT1ARs [71, 74, 88]. ineffective high dose of CBD as anxiolytic in the EPM [66]. Given TRPV1 receptors have anxiogenic effects, this may Contextual Fear Conditioning, Fear Extinction, indicate that at higher doses, CBD’s interaction with TRPV1 and Reconsolidation Blockade receptors to some extent impedes anxiolytic actions, although was notably not sufficient to produce anxiogenic Several studies assessed CBD using contextual fear condition- effects. ing. Briefly, this paradigm involves pairing a neutral context, the conditioned stimulus (CS), with an aversive unconditioned Stress-induced Anxiety Models stimulus (US), a mild foot shock. After repeated pairings, the subject learns that the CS predicts the US, and subsequent CS Stress is an important contributor to anxiety disorders, and presentation elicits freezing and other physiological re- traumatic stress exposure is essential to the development of sponses. Systemic administration of CBD prior to CS PTSD. Systemically administered CBD reduced acute in- re-exposure reduced conditioned cardiovascular re- creases in heart rate and blood pressure induced by restraint sponses [63], an effect reproduced by microinjection of stress, as well as the delayed (24 h) anxiogenic effects of stress CBD into the BNST, and partially mediated by 5- in the EPM, partially by 5-HT1AR activation [67, 73]. How- HT1AR activation [79]. Similarly, CBD in the prelimbic ever intra-BNST microinjection of CBD augmented stress- cortex reduced conditioned freezing [70], an effect induced heart rate increase, also partially via 5-HT1ARactiva- preventedby5-HT1ARblockade[87]. By contrast, tion [85]. In a subchronic study, CBD administered daily 1 h CBD microinjection in the infralimbic cortex enhanced after predator stress (a proposed model of PTSD) reduced the conditionedfreezing[70]. Finally, El Batsh et al. [80] long-lasting anxiogenic effects of chronic predator stress, par- reported that repeated CBD doses over 21 days, that is tially via 5-HT1AR activation [77]. In a chronic study, system- chronic as opposed to acute treatment, facilitated condi- ic CBD prevented increased anxiety produced by chronic un- tioned freezing. In this study, CBD was administered predictable stress, in addition to increasing hippocampal prior to conditioning rather than prior to re-exposure

AEA; these anxiolytic effects depended upon CB1R activation as in acute studies, thus further directly comparable and hippocampal neurogenesis, as demonstrated by genetic studies are required. ablation techniques [81]. Prior stress also appears to modulate CBD has also been shown to enhance extinction of CBD’s anxiogenic effects: microinjection of CBD into the contextually conditioned fear responses. Extinction train- prelimbic cortex of unstressed animals was anxiogenic in the ing involves repeated CS exposure in the absence of the EPM but following restraint stress was found to be anxiolytic US, leading to the formation of a new memory that [87]. Likewise, systemic CBD was anxiolytic in the EPM inhibits fear responses and a decline in freezing over following but not prior to stress [65]. subsequent training sessions. Systemic CBD administra- tion immediately before training markedly enhanced ex-

PD and Compulsive Behavior Models tinction, and this effect depended on CB1R activation, without involvement of TRPV1 receptors [65]. Further

CBD inhibited escape responses in the ETM and increased studies showed CB1Rs in the infralimbic cortex may be DPAG escape electrical threshold [68], both proposed models involved in this effect [82]. of panic attacks [95]. These effects partially depended on 5- CBD also blocked reconsolidation of aversive memo-

HT1AR activation but were not affected by CB1R blockade. ries in rat [76]. Briefly, fear memories, when reactivated CBD was also panicolytic in the predator–prey model, which by re-exposure (retrieval), enter into a labile state in Cannabidiol as a Potential Treatment for Anxiety Disorders 831 which the memory trace may either be reconsolidated or Human Experimental and Clinical Studies extinguished [97], and this process may be pharmacolog- ically modulated to achieve reconsolidation blockade or Evidence from Acute Psychological Studies extinction. When administered immediately following re- trieval, CBD prevented freezing to the conditioned con- Relevant studies are summarized in Table 2. The anxiolytic text upon further re-exposure, and no reinstatement or effects of CBD in humans were first demonstrated in the con- spontaneous recovery was observed over 3 weeks, con- text of reversing the anxiogenic effects of THC. CBD reduced sistent with reconsolidation blockade rather than extinc- THC-induced anxiety when administered simultaneously with tion [76]. This effect depended on CB1R activation but this agent, but had no effect on baseline anxiety when admin- not 5-HT1ARactivation[76]. istered alone [99, 100]. Further studies using higher doses supported a lack of anxiolytic effects at baseline [101, 107]. Summary and Clinical Relevance By contrast, CBD potently reduces experimentally induced anxiety or fear. CBD reduced anxiety associated with a simu- Overall, existing preclinical evidence strongly supports lated public speaking test in healthy subjects, and in subjects the potential of CBD as a treatment for anxiety disor- with SAD, showing a comparable efficacy to ipsapirone (a 5- ders. CBD exhibits a broad range of actions, relevant to HT1ARagonist)ordiazepam[98, 105]. CBD also reduced the multiple symptom domains, including anxiolytic, presumed anticipatory anxiety associated with undergoing a panicolytic, and anticompulsive actions, as well as a single-photon emission computed tomography (SPECT) im- decrease in autonomic arousal, a decrease aging procedure, in both healthy and SAD subjects [102, 104]. in conditioned fear expression, enhancement of fear ex- Finally, CBD enhanced extinction of fear memories in healthy tinction, reconsolidation blockade, and prevention of the volunteers: specifically, inhaled CBD administered prior to or long-term anxiogenic effects of stress. Activation of 5- after extinction training in a contextual fear conditioning par-

HT1ARs appears to mediate anxiolytic and panicolytic adigm led to a trend-level enhancement in the reduction of effects, in addition to reducing conditioned fear expres- skin conductance response during reinstatement, and a signif- sion, although CB1R activation may play a limited role. icant reduction in expectancy (of shock) ratings during rein- By contrast, CB1R activation appears to mediate CBD’s statement [106]. anticompulsive effects, enhancement of fear extinction, reconsolidation blockade, and capacity to prevent the Evidence from Neuroimaging Studies long-term anxiogenic consequences of stress, with in- volvement of hippocampal neurogenesis. Relevant studies are summarized in Table 3. In a SPECTstudy While CBD predominantly has acute anxiolytic ef- of resting cerebral blood flow (rCBF) in normal subjects, fects, some species discrepancies are apparent. In addi- CBD reduced rCBF in left medial temporal areas, including tion, effects may be contingent on prior stress and vary the amygdala and hippocampus, as well as the hypothalamus according to brain region. A notable contrast between and left posterior cingulate gyrus, but increased rCBF in the CBD and other agents that target the eCB system, in- left parahippocampal gyrus. These rCBF changes were not cluding THC, direct CB1R agonists and FAAH inhibi- correlated with anxiolytic effects [102]. In a SPECT study, tors, is a lack of anxiogenic effects at a higher dose. by the same authors, in patients with SAD, CBD reduced Further receptor-specific studies may elucidate the recep- rCBF in overlapping, but distinct, limbic and paralimbic areas; tor specific basis of this distinct dose response profile. again, with no correlations to anxiolytic effects [104]. Further studies are also required to establish the efficacy In a series of placebo-controlled studies involving 15 of CBD when administered in chronic dosing, as rela- healthy volunteers, Fusar-Poli et al. investigated the effects tively few relevant studies exist, with mixed results, in- of CBD and THC on task-related blood-oxygen-level depen- cluding both anxiolytic and anxiogenic outcomes. dent functional magnetic resonance imaging activation, spe- Overall, preclinical evidence supports systemic CBD cifically the go/no-go and fearful faces tasks [109, 110]. The as an acute treatment of GAD, SAD, PD, OCD, and go/no-go task measures response inhibition, and is associated PTSD, and suggests that CBD has the advantage of with activation of medial prefrontal, dorsolateral prefrontal, not producing anxiogenic effects at higher dose, as dis- and parietal areas [111]. Response activation is diminished tinct from other agents that enhance CB1R activation. In in PTSD and other anxiety disorders, and increased activation particular, results show potential for the treatment of predicts response to treatment [112]. CBD produced no multiple PTSD symptom domains, including reducing changes in predicted areas (relative to placebo) but reduced arousal and avoidance, preventing the long-term adverse activation in the left insula, superior temporal gyrus, and trans- effects of stress, as well as enhancing the extinction and verse temporal gyrus. The fearful faces task activates the blocking the reconsolidation of persistent fear memories. amygdala, and other medial temporal areas involved in 832 Blessing et al.

Table 2 Human psychological studies

Study Subjects, CBD route, Measure Effect design dose

Karniol et al. [99]HV, Oral, 15, 30, 60 mg, alone Anxiety and pulse rate after ↓ THC-induced increases in DBP or with THC, THC and at baseline subjective anxiety and acute, at 55, 95, 155, and pulse rate 185 min No effect at baseline Zuardi et al., [100]HV, Oral 1 mg/kg alone or with STAI score after THC ↓ THC-induced increases in DBP THC, acute, 80 min STAI scores Zuardi et al. [98]HV, Oral 300 mg, VAMS, STAI and BP ↓ STAI scores DBP acute, 80 min following SPST ↓ VA MS s cor es ↓ BP Martin-Santos et al. [101]HV, Oral 600 mg, Baseline anxiety and No effect DBP acute, 1, 2, 3 h pulse rate Crippa et al. [102]10HV, Oral 400 mg, VAMS before SPECT ↓ VA MS s cor es DBP acute, 60 and 75 min SPECT Bhattacharyya et al. [103]15HV Oral 600 mg, STAI scores ↓ STAI scores DBP acute, 1, 2, 3 h VAMS scores ↓ VA MS s cor es Crippa et al. [104] SAD and HC Oral 400 mg, VAMS before SPECT ↓ VA MS s cor es DBP acute, 75 and 140 min SPECT Bergamaschi et al. [105] SAD and HC DBP Oral 600 mg, acute, VAMS, SSPS-N, cognitive ↓ VAMS, SSPS-N and cognitive 1, 2, 3 h impairment, SCR, HR impairment, no effect on SCR after SPST or HR Das et al. [106]HV Inhaled, 32 mg, acute, SCR and shock expectancy CBD after extinction training DBP immediately following, following extinction produced trend level reduction before, after extinction inSCRanddecreasedshock expectancy Hindocha et al. [107] Varying in schizotypy and Inhaled, 16 mg, acute Baseline VAS anxiety No significant effect of CBD cannabis use, DBP

HV = healthy volunteers; DBP = double-blind placebo; SAD = social anxiety disorder; HC = healthy controls; THC = Δ 9-tetrahydrocannabinol; STAI = Spielberger’s state trait anxiety inventory; VAMS = visual analog mood scale; BP = blood pressure; SPST = simulated public speaking test; SCR = skin conductance response; SPECT = single-photon emission computed tomography; SSPS-N = negative self-evaluation subscale; HR = heart rate; VAS = visual analog scale, CBD = cannabidiol

Table 3 Neuroimaging studies

Study Subjects, design CBD route, dose, timing Measure Effect of CBD

Crippa et al. [102]10HV, Oral 400 mg, SPECT, resting (rCBF) ↓ rCBF in left medial temporal cluster, DBP acute, 60 and 75 min including amygdala and HPC, also ↓ rCBF in the HYP and posterior cingulate gyrus ↑ rCBF in left PHG Borgwardt et al. [108]15HV, Oral 600 mg, fMRI during oddball and ↓ Activation in left insula, STG and MTG DBP acute, 1–2h go/no-go task Fusar-Poli et al. [109]15HV, Oral 600 mg, fMRI activation during ↓ Activation in left medial temporal region, DBP acute, 1–2h fearful faces task including amygdala and anterior PHG, and in right ACC and PCC Fusar-Poli et al. [110]15HV, Oral 600 mg, fMRI functional connectivity ↓ Functional connectivity between L) AMY DBP acute, 1–2h during fearful faces task and ACC Crippa et al. [104] SAD and HC Oral 400 mg, SPECT, resting (rCBF) ↓ rCBF in the left PHG, HPC and ITG. DBP acute, 75 and 140 min ↑ rCBF in the right posterior cingulate gyrus

CBD = cannabidiol; HV = healthy controls; DBP = double-blind placebo; SAD = social anxiety disorder; HC = healthy controls; SPECT = single-photo emission computed tomography; rCBF = regional cerebral blood flow; fMRI = functional magnetic resonance imaging; HPC = hippocampus; HYP = hypothalamus; PHG = parahippocampal gyrus; STG = superior temporal gyrus; MTG = medial temporal gyrus; ACC = anterior cingulate cortex; PCC = posterior cingulate cortex Cannabidiol as a Potential Treatment for Anxiety Disorders 833 emotion processing, and heightened amygdala response acti- Conclusions vation has been reported in anxiety disorders, including GAD and PTSD [113, 114]. CBD attenuated blood-oxygen-level Preclinical evidence conclusively demonstrates CBD’seffica- dependent activation in the left amygdala, and the anterior cy in reducing anxiety behaviors relevant to multiple disor- and posterior cingulate cortex in response to intensely fearful ders, including PTSD, GAD, PD, OCD, and SAD, with a faces, and also reduced amplitude in skin conductance fluctu- notable lack of anxiogenic effects. CBD’s anxiolytic actions ation, which was highly correlated with amygdala activation appear to depend upon CB1Rs and 5-HT1ARs in several brain [109]. Dynamic causal modeling analysis in this data set fur- regions; however, investigation of additional receptor actions ther showed CBD reduced forward functional connectivity may reveal further mechanisms. Human experimental find- between the amygdala and anterior cingulate cortex [110]. ings support preclinical findings, and also suggest a lack of anxiogenic effects, minimal sedative effects, and an excellent safety profile. Current preclinical and human findings mostly Evidence from Epidemiological and Chronic Studies involve acute CBD dosing in healthy subjects, so further stud- ies are required to establish whether chronic dosing of CBD Epidemiological studies of various neuropsychiatric disorders has similar effects in relevant clinical populations. Overall, indicate that a higher CBD content in chronically consumed this review emphasizes the potential value and need for further cannabis may protect against adverse effects of THC, includ- study of CBD in the treatment of anxiety disorders. ing psychotic symptoms, drug cravings, memory loss, and hippocampal gray matter loss [115–118] (reviewed in [119]). As THC acutely induces anxiety, this pattern may also be Required Author Forms Disclosure forms provided by the authors are evident for chronic anxiety symptoms. Two studies were iden- available with the online version of this article. tified, including an uncontrolled retrospective study in civilian patients with PTSD patients [120], and a case study in a pa- tient with severe sexual abuse-related PTSD [121], which References showed that chronic cannabis use significantly reduces PTSD symptoms; however, these studies did not include data on the 1. Kroenke K, Spitzer RL, Williams JB, Monahan PO, Lowe B. THC:CBD ratio. Thus, overall, no outcome data are currently Anxiety disorders in primary care: prevalence, impairment, co- morbidity, and detection. Ann Intern Med 2007;146:317-325. available regarding the chronic effects of CBD in the treat- 2. Khan A, Leventhal RM, Khan S, Brown WA. Suicide risk in ment of anxiety symptoms, nor do any data exist regarding the patients with anxiety disorders: a meta-analysis of the FDA data- potential protective effects of CBD on anxiety potentially in- base. J Affect Disord 2002;68:183-190. duced by chronic THC use. 3. Olatunji BO, Cisler JM, Tolin DF. Quality of life in the anxiety dis- orders: a meta-analytic review. Clin Psychol Rev 2007;27:572-581. 4. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Summary and Clinical Relevance Walters EE. Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 2005;62:593-602. Evidence from human studies strongly supports the potential 5. Wang PS, Lane M, Olfson M, Pincus HA, Wells KB, Kessler RC. for CBD as a treatment for anxiety disorders: at oral doses Twelve-month use of mental health services in the United States: ranging from 300 to 600 mg, CBD reduces experimentally results from the National Comorbidity Survey Replication. Arch induced anxiety in healthy controls, without affecting baseline Gen Psychiatry 2005;62:629-640. 6. Gustavsson A, Svensson M, Jacobi F, et al. Cost of disorders of the anxiety levels, and reduces anxiety in patients with SAD. brain in Europe 2010. Eur Neuropsychopharmacol 2011;21:718-779. Limited results in healthy subjects also support the efficacy 7. Otto MW, Tuby KS, Gould RA, McLean RY, Pollack MH. An of CBD in acutely enhancing fear extinction, suggesting po- effect-size analysis of the relative efficacy and tolerability of sero- tential for the treatment of PTSD, or for enhancing cognitive tonin selective reuptake inhibitors for panic disorder. Am J Psychiary 2001;158:1989-1992. behavioral therapy. Neuroimaging findings provide evidence 8. Ballenger JC. Remission rates in patients with anxiety disorders of neurobiological targets that may underlie CBD’s anxiolytic treated with paroxetine. J Clin Psychiatry 2004;65:1696-1707. effects, including reduced amygdala activation and altered 9. Krystal JH, Rosenheck RA, Cramer JA, et al. Adjunctive risperi- medial prefrontal amygdala connectivity, although current done treatment for antidepressant-resistant symptoms of chronic military service-related PTSD: a randomized trial. JAMA findings are limited by small sample sizes, and a lack of inde- 2011;306:493-502. pendent replication. Further studies are also required to estab- 10. Shin HJ, Greenbaum MA, Jain S, Rosen CS. Associations of lish whether chronic, in addition to acute CBD dosing is an- psychotherapy dose and SSRI or SNRI refills with mental health xiolytic in human. Also, clinical findings are currently limited outcomes among veterans with PTSD. Psychiatr Serv 2014;65: 1244-1248. to SAD, whereas preclinical evidence suggests CBD’spoten- 11. Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non- tial to treat multiple symptom domains relevant to GAD, PD, psychotropic plant cannabinoids: new therapeutic opportunities and, particularly, PTSD. from an ancient herb. Trends Pharmacol Sci 2009;30:515-527. 834 Blessing et al.

12. Campos AC, Moreira FA, Gomes FV,Del Bel EA, Guimaraes FS. 34. Llorente-Berzal A, Terzian AL, di Marzo V, Micale V, Viveros Multiple mechanisms involved in the large-spectrum therapeutic MP, Wotjak CT. 2-AG promotes the expression of conditioned potential of cannabidiol in psychiatric disorders. Philos Trans R fear via cannabinoid receptor type 1 on GABAergic neurons. Soc Lond Ser B Biol Sci 2012;367:3364-3378. Psychopharmacology 2015;232: 2811-2825. 13. Schier ARD, Ribeiro NP, Silva AC, et al. Cannabidiol, a Cannabis 35. Marsicano G, Wotjak CT, Azad SC, et al. The endogenous canna- sativa constituent, as an anxiolytic drug. Rev Bras Psiquiatr binoid system controls extinction of aversive memories. Nature 2012;34:S104-S117. 2002;418:530-534. 14. Schier ARD, de Oliveira Ribeiro NP, Coutinho DS, et al. 36. Dincheva I, Drysdale AT, Hartley CA. FAAH genetic variation Antidepressant-like and anxiolytic-like effects of cannabidiol: A enhances fronto-amygdala function in mouse and human. Nat chemical compound of Cannabis sativa. CNS Neurol Disord Commun 2015;6:6395. Drug Targets 2014;13:953-960. 37. Gray JM, Vecchiarelli HA, Morena M, et al. Corticotropin- 15. Micale V, Di Marzo V, Sulcova A, Wotjak CT, Drago F. releasing hormone drives anandamide hydrolysis in the amygdala Endocannabinoid system and mood disorders: priming a target to promote anxiety. J Neurosci 2015;35:3879-3892. for new therapies. Pharmacol Ther 2013;138:18-37. 38. Evanson NK, Tasker JG, Hill MN, Hillard CJ, Herman JP. Fast 16. Mechoulam R, Peters M, Murillo-Rodriguez E, Hanus LO. feedback inhibition of the HPA axis by glucocorticoids is mediated — Cannabidiol recent advances. Chem Biodivers 2007;4:1678- by endocannabinoid signaling. Endocrinology 2010;151:4811-4819. 1692. 39. Abush H, Akirav I. Cannabinoids ameliorate impairments induced 17. Marco EM, Garcia-Gutierrez MS, Bermudez-Silva FJ, et al. by chronic stress to synaptic plasticity and short-term memory. Endocannabinoid system and psychiatry: in search of a neurobio- Neuropsychopharmacology 2013;38:1521-1534. logical basis for detrimental and potential therapeutic effects. Front Behav Neurosci 2011;5:63. 40. Hill MN, Patel S, Carrier EJ, et al. Downregulation of 18. Devinsky O, Cilio MR, Cross H, et al. Cannabidiol: endocannabinoid signaling in the hippocampus following chronic unpredictable stress. Neuropsychopharmacology 2005;30;508-515. Pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia 2014;55:791-802. 41. Qin Z, Zhou X, Pandey NR, et al. Chronic stress induces anxiety 19. Robson PJ, Guy GW, Di Marzo V.Cannabinoids and schizophrenia: via an amygdalar intracellular cascade that impairs therapeutic prospects. Curr Pharm Design 2014;20:2194-2204. endocannabinoid signaling. Neuron 2015;85:1319-1331. 20. Bergamaschi MM, Queiroz RH, Zuardi AW, Crippa JA. Safety 42. Neumeister A. The endocannabinoid system provides an avenue and side effects of cannabidiol, a Cannabis sativa constituent. for evidence-based treatment development for PTSD. Depress Curr Drug Saf 2011;6: 237-249. Anxiety 2013;30:93-96. 21. McPartland JM, Duncan M, Di Marzo V, Pertwee RG. Are 43. Papini S, Sullivan GM, Hien DA, Shvil E, Neria Y. Toward a cannabidiol and Delta(9) -tetrahydrocannabivarin negative modu- translational approach to targeting the endocannabinoid system lators of the endocannabinoid system? A systematic review. Br J in posttraumatic stress disorder: a critical review of preclinical Pharmacol 2015;172:737-753. research. Biol Psychol 2015;104:8-18. 22. Di Marzo V, Bisogno T, De Petrocellis L. Anandamide: Some like 44. Ragen BJ, Seidel J, Chollak C, Pietrzak RH, Neumeister A. it hot. Trends Pharmacol Sci 2001;22:346-349. Investigational drugs under development for the treatment of 23. Wilson RI, Nicoll RA. Endocannabinoid signaling in the brain. PTSD. Exp Opin Invest Drugs 2015;24:659-672. Science 2002;296: 678-682. 45. Viveros MP, Marco EM, File SE. Endocannabinoid system and stress 24. Battista N, Di Tommaso M, Bari M, Maccarrone M. The and anxiety responses. Pharmacol Biochem Behav 2005;81:331-342. endocannabinoid system: An overview. Front Behav Neurosci 46. Rubino T, Realini N, Castiglioni C, et al. Role in anxiety behavior 2012;6:9. of the endocannabinoid system in the prefrontal cortex. Cereb 25. Lee SH, et al. Multiple forms of endocannabinoid and Cortex 2008;18:1292-1301. endovanilloid signaling regulate the tonic control of GABA re- 47. Moreira FA, Aguiar DC, Terzian AL, Guimaraes FS, Wotjak CT. lease. J Neurosci 2015;35:10039-10057. Cannabinoid type 1 receptors and transient receptor potential 26. Kauer JA, Gibson HE. Hot flash: TRPV channels in the brain. vanilloid type 1 channels in fear and anxiety-two sides of one Trends Neurosci 2009;32:215-224. coin? Neuroscience 2012;204:186-192. 27. Aguiar DC, Moreira FA, Terzian AL, et al. Modulation of defen- 48. Bisogno T, Hanus L, De Petrocellis L, et al. Molecular targets for sive behavior by transient receptor potential vanilloid type-1 cannabidiol and its synthetic analogues: effect on vanilloid VR1 (TRPV1) channels. Neurosci Biobehav Rev 2014;46:418-428. receptors and on the cellular uptake and enzymatic hydrolysis of 28. Silvestri C, Di Marzo V. The endocannabinoid system in energy anandamide. Br J Pharmacol 2001;134:845-852. homeostasis and the etiopathology of metabolic disorders. Cell 49. Haller J, et al. Interactions between environmental aversiveness and Metab 2013;17:475-490. the anxiolytic effects of enhanced cannabinoid signaling by FAAH 29. Castillo PE, Younts TJ, Chavez AE, Hashimotodani Y. inhibition in rats. Psychopharmacology 2009;204:607-616. Endocannabinoid signaling and synaptic function. Neuron 50. Chessick CA, Allen MH, Thase M, et al. Azapirones for general- 2012;76:70-81. ized anxiety disorder. Cochrane Database Syst Rev 30. Riebe CJ, Pamplona FA, Kamprath K, Wotjak CT. Fear relief- 2006;CD006115. toward a new conceptual frame work and what endocannabinoids gotta do with it. Neuroscience 2012;204:159-185. 51. Roncon CM, Biesdorf C, Coimbra NC, et al. Cooperative regula- 31. McLaughlin RJ, Hill MN, Gorzalka BB. A critical role for tion of anxiety and panic-related defensive behaviors in the rat prefrontocortical endocannabinoid signaling in the regulation of periaqueductal grey matter by 5-HT1A and mu-receptors. J stress and emotional behavior. Neurosci Biobehav Rev 2014;42: Psychopharmacol 2013;27:1141-1148. 116-131. 52. Zhou J, Cao X, Mar AC, et al. Activation of postsynaptic 5-HT1A 32. Moreira FA, Lutz B. The endocannabinoid system: emotion, receptors improve stress adaptation. Psychopharmacology learning and addiction. Addict Biol 2008;13:196-212. 2014;231:2067-2075. 33. Ruehle S, Rey AA, Remmers F, Lutz B. The endocannabinoid 53. Saito Y, Matsumoto M, Yanagawa Y, et al. Facilitation of fear extinc- system in anxiety, fear memory and habituation. J tion by the 5-HT(1A) receptor agonist tandospirone: possible in- Psychopharmacol 2012;26:23-39. volvement of dopaminergic modulation. Synapse 2013;67:161-170. Cannabidiol as a Potential Treatment for Anxiety Disorders 835

54. Sprouse JS, Aghajanian GK. Electrophysiological responses of 72. Gomes FV, Resstel LB, Guimaraes FS. The anxiolytic-like effects serotoninergic dorsal raphe neurons to 5-HT1A and 5-HT1B ag- of cannabidiol injected into the bed nucleus of the stria terminalis onists. Synapse 1987;1:3-9. are mediated by 5-HT1A receptors. Psychopharmacology (Berl) 55. Sun YN, Wang T, Wang Y, et al. Activation of 5-HT receptors in 2011;213:465-473. the medial subdivision of the central nucleus of the amygdala 73. Granjeiro EM, Gomes FV,Guimaraes FS, Correa FM, Resstel LB. produces anxiolytic effects in a rat model of Parkinson's disease. Effects of intracisternal administration of cannabidiol on the car- Neuropharmacology 2015;95:181-191. diovascular and behavioral responses to acute restraint stress. 56. Celada P, Bortolozzi A, Artigas F. Serotonin 5-HT1A receptors as Pharmacol Biochem Behav 2011;99:743-748. targets for agents to treat psychiatric disorders: rationale and cur- 74. Deiana S, Watanabe A, Yamasaki Y. Plasma and brain pharmacoki- rent status of research. CNS Drugs 2013;27:703-716. netic profile of cannabidiol (CBD), cannabidivarine (CBDV), 57. Russo EB, Burnett A, Hall B, Parker KK. Agonistic properties of Delta(9)-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) cannabidiol at 5-HT1a receptors. Neurochem Res 2005;30:1037- in rats and mice following oral and intraperitoneal administration 1043. and CBD action on obsessive-compulsive behaviour. 58. Rock EM, Bolognini D, Limebeer CL, et al. Cannabidiol, a non- Psychopharmacology (Berl) 2012;219:859-873. psychotropic component of cannabis, attenuates vomiting and 75. Uribe-Marino A, et al. Anti-aversive effects of cannabidiol on nausea-like behaviour via indirect agonism of 5-HT(1A) innate fear-induced behaviors evoked by an ethological model of somatodendritic autoreceptors in the dorsal raphe nucleus. Br J panic attacks based on a prey vs the wild snake Epicrates cenchria Pharmacol 2012;165:2620-2634. crassus confrontation paradigm. Neuropsychopharmacology 59. Silveira Filho NG, Tufik S. Comparative effects between 2012;37:412-421. cannabidiol and diazepam on neophobia, food intake and conflict 76. Stern CA, Gazarini L, Takahashi RN, Guimaraes FS, Bertoglio LJ. behavior. Res Commun Psychol Psychiatry Behav 1981;6:25-26. On disruption of fear memory by reconsolidation blockade: evi- 60. Zuardi AW, Finkelfarb E, Bueno OF, Musty RE, Karniol IG. dence from cannabidiol treatment. Neuropsychopharmacology Characteristics of the stimulus produced by the mixture of 2012;37:2132-2142. cannabidiol with delta 9-tetrahydrocannabinol. Arch Int 77. Campos AC, Ferreira FR, Guimaraes FS. Cannabidiol blocks Pharmacodyn Ther 1981;249:137-146. long-lasting behavioral consequences of predator threat stress: 61. Onaivi ES, Green MR, Martin BR. Pharmacological characteriza- possible involvement of 5HT1A receptors. J Psychiatr Res tion of cannabinoids in the elevated plus maze. J Pharmacol Exp 2012;46:1501-1510. Ther 1990;253:1002-1009. 78. Hsiao YT, Yi PL, Li CL, Chang FC. Effect of cannabidiol on sleep disruption induced by the repeated combination tests consisting of 62. Moreira FA, Aguiar DC, Guimaraes FS. Anxiolytic-like ef- open field and elevated plus-maze in rats. Neuropharmacology fect of cannabidiol in the rat Vogel conflict test. Prog 2012;62:373-384. Neuropsychopharmacol Biol Psychiatry 2006;30:1466-1471. 79. Gomes FV, Resstel LB, Guimaraes FS, et al. Cannabidiol injected 63. Resstel LB, Joca SR, Moreira FA, Correa FM, Guimaraes FS. into the bed nucleus of the stria terminalis reduces the expression Effects of cannabidiol and diazepam on behavioral and cardiovas- of contextual fear conditioning via 5-HT1A receptors. J cular responses induced by contextual conditioned fear in rats. Psychopharmacol 2012;26:104-113. Behav Brain Res 2006;172:294-298. 80. El Batsh MM, Assareh N, Marsden CA, Kendall DA. Anxiogenic- 64. Campos AC, Guimaraes FS. Involvement of 5HT1A receptors in like effects of chronic cannabidiol administration in rats. the anxiolytic-like effects of cannabidiol injected into the dorso- Psychopharmacology (Berl) 2012;221:239-247. lateral periaqueductal gray of rats. Psychopharmacology (Berl) 81. Campos AC, Ortega Z, Palazuelos J, et al. The anxiolytic effect of 2008;199:223-230. cannabidiol on chronically stressed mice depends on hippocampal 65. Bitencourt RM, Pamplona FA, Takahashi RN. Facilitation of con- neurogenesis: involvement of the endocannabinoid system. Int J textual fear memory extinction and anti-anxiogenic effects of Neuropsychopharmacol 2013;16:1407-1419. AM404 and cannabidiol in conditioned rats. Eur 82. Do Monte FH, Souza RR, Bitencourt RM, Kroon JA, Takahashi Neuropsychopharmacol 2009;18:849-859. RN. Infusion of cannabidiol into infralimbic cortex facilitates fear 66. Campos AC, Guimaraes FS. Evidence for a potential role for extinction via CB1 receptors. Behav Brain Res 2013;250:23-27. TRPV1 receptors in the dorsolateral periaqueductal gray in the 83. Campos AC, de Paula Soares V, Carvalho MC, et al. Involvement attenuation of the anxiolytic effects of cannabinoids. Prog of serotonin-mediated neurotransmission in the dorsal Neuropsychopharmacol Biol Psychiatry 2009;33:1517-1521. periaqueductal gray matter on cannabidiol chronic effects in 67. Resstel LB, Tavares RF, Lisboa SF, et al. 5-HT1A receptors are panic-like responses in rats. Psychopharmacology (Berl) involved in the cannabidiol-induced attenuation of behavioural 2013;226:13-24. and cardiovascular responses to acute restraint stress in rats. Br J 84. Almeida V, Levin R, Peres FF, et al. Cannabidiol exhibits anxio- Pharmacol 2009;156:181-188. lytic but not antipsychotic property evaluated in the social inter- 68. Soares Vde P, Campos AC, Bortoli VC, et al. Intra-dorsal action test. Prog Neuropsychopharmacol Biol Psychiatry 2013;41: periaqueductal gray administration of cannabidiol blocks panic- 30-35. like response by activating 5-HT1A receptors. Behav Brain Res 85. Gomes FV, Alves FH, Guimaraes FS, et al. Cannabidiol administra- 2010;213:225-229. tion into the bed nucleus of the stria terminalis alters cardiovascular 69. Long LE, Chesworth R, Huang XF. A behavioural comparison of responses induced by acute restraint stress through 5-HT(1)A recep- acute and chronic Delta9-tetrahydrocannabinol and cannabidiol in tor. Eur Neuropsychopharmacol 2013;23:1096-1104. C57BL/6JArc mice. Int J Neuropsychopharmacol 2010;13:861-876. 86. Twardowschy A, Castiblanco-Urbina MA, Uribe-Marino A, et al. 70. Lemos JI, Resstel LB, Guimaraes FS. Involvement of the The role of 5-HT1A receptors in the anti-aversive effects of prelimbic prefrontal cortex on cannabidiol-induced attenuation cannabidiol on panic attack-like behaviors evoked in the presence of contextual conditioned fear in rats. Behav Brain Res of the wild snake Epicrates cenchria crassus (Reptilia, Boidae). J 2010;207:105-111. Psychopharmacol 2013;27:1149-1159. 71. Casarotto PC, Gomes FV,Resstel LB, Guimaraes FS. Cannabidiol 87. Fogaca MV, Reis FM, Campos AC, Guimaraes FS. Effects of inhibitory effect on marble-burying behaviour: involvement of intra-prelimbic prefrontal cortex injection of cannabidiol on CB1 receptors. Behav Pharmacol 2010;21:353-358. anxiety-like behavior: involvement of 5HT1A receptors and 836 Blessing et al.

previous stressful experience. Eur Neuropsychopharmacol 105. Bergamaschi MM, Queiroz RH, Chagas MH, et al. Cannabidiol 2014;24:410-419. reduces the anxiety induced by simulated public speaking in 88. Nardo M, Casarotto PC, Gomes FV, Guimaraes FS. Cannabidiol treatment-naive social phobia patients. reverses the mCPP-induced increase in marble-burying behavior. Neuropsychopharmacology 2011;36:1219-1226. Fundam Clin Pharmacol 2014;28:544-550. 106. Das RK, Kamboj SK, Ramadas M, et al. Cannabidiol enhances 89. da Silva JA, Biagioni AF, Almada RC, et al. Dissociation between consolidation of explicit fear extinction in humans. the panicolytic effect of cannabidiol microinjectedintothe Psychopharmacology 2013;226:781-792. substantia nigra, pars reticulata, and fear-induced antinociception 107. Hindocha C, Freeman TP, Schafer G, et al. Acute effects of delta- elicited by bicuculline administration in deep layers of the superior 9-tetrahydrocannabinol, cannabidiol and their combination on fa- colliculus: The role of CB-cannabinoid receptor in the ventral cial emotion recognition: a randomised, double-blind, placebo- mesencephalon. Eur J Pharmacol 2015;758:153-163. controlled study in cannabis users. Eur Neuropsychopharmacol 90. Guimaraes FS, Chiaretti TM, Graeff FG, Zuardi AW. Antianxiety 2015;25:325-334. effect of cannabidiol in the elevated plus-maze. 108. Borgwardt SJ, Allen P, Bhattacharyya S, et al. Neural basis of Psychopharmacology (Berl) 1990;100:558-559. Delta-9-tetrahydrocannabinol and cannabidiol: effects during re- 91. Bandler R, Shipley MT. Columnar organization in the midbrain sponse inhibition. Biol Psychiatry 2008;64:966-973. periaqueductal gray: modules for emotional expression? Trends 109. Fusar-Poli P, Crippa JA, Bhattacharyya S. Distinct effects of {del- Neurosci 1994;17:379-389. ta}9-tetrahydrocannabinol and cannabidiol on neural activation 92. Nashold BS, Jr, Wilson WP, Slaughter DG. Sensations evoked by during emotional processing. Arch Gen Psychiatry 2009;66:95- stimulation in the midbrain of man. J Neurosurg 1969;30:14-24. 105. 93. Walker DL, Miles LA, Davis M. Selective participation of the bed 110. Fusar-Poli P, Allen P, Bhattacharyya S. Modulation of effective nucleus of the stria terminalis and CRF in sustained anxiety-like connectivity during emotional processing by Delta 9- versus phasic fear-like responses. Prog Neuropsychopharmacol tetrahydrocannabinol and cannabidiol. Int J Biol Psychiatry 2009;33:1291-1308. Neuropsychopharmacol 2010;13:421-432. 94. Sierra-Mercado D, Padilla-Coreano N, Quirk GJ. Dissociable 111. Rubia K, Russell T, Overmeyer S, et al. Mapping motor inhibition: roles of prelimbic and infralimbic cortices, ventral hippocampus, conjunctive brain activations across different versions of go/no-go and basolateral amygdala in the expression and extinction of con- and stop tasks. Neuroimage 2001;13:250-261. ditioned fear. Neuropsychopharmacology 2011;36:529-538. 112. Falconer E, Allen A, Felmingham KL, Williams LM, Bryant RA. 95. Schenberg LC, Bittencourt AS, Sudre EC, Vargas LC. Modeling Inhibitory neural activity predicts response to cognitive-behavioral panic attacks. Neurosci Biobehav Rev 2001;25;647-659. therapy for posttraumatic stress disorder. J Clin Psychiatry 96. Thomas A, Burant A, Bui N, Graham D, Yuva-Paylor LA, 2013;74:895-901. Paylor R. Marble burying reflects a repetitive and persev- 113. Mochcovitch MD, da Rocha Freire RC, Garcia RF, Nardi AE. A erative behavior more than novelty-induced anxiety. systematic review of fMRI studies in generalized anxiety disorder: Psychopharmacology (Berl) 2009;204;361-373. evaluating its neural and cognitive basis. J Affect Disord 97. Suzuki A, Josselyn SA, Frankland PW, et al. Memory 2014;167:336-342. reconsolidation and extinction have distinct temporal and bio- 114. Patel R, Spreng RN, Shin LM, Girard TA. Neurocircuitry models chemical signatures. J Neurosci 2004;24:4787-4795. of posttraumatic stress disorder and beyond: a meta-analysis of 98. Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG. Action of functional neuroimaging studies. Neurosci Biobehav Rev cannabidiol on the anxiety and other effects produced by delta 2012;36:2130-2142. 9-THC in normal subjects. Psychopharmacology (Berl) 1982;76: 115. Morgan CJ, Freeman TP, Schafer GL, Curran HV. Cannabidiol atten- 245-250. uates the appetitive effects of Delta 9-tetrahydrocannabinol in humans 99. Karniol IG, Shirakawa I, Kasinski N, Pfeferman A, Carlini EA. smoking their chosen cannabis. Neuropsychopharmacology 2010;35: Cannabidiol interferes with the effects of delta 9 - tetrahydrocan- 1879-1885. nabinol in man. Eur J Pharmacol 1974;28:172-177. 116. Morgan CJ, Curran HV. Effects of cannabidiol on schizophrenia- 100. Zuardi AW, Cosme RA, Graeff FG, Guimaraes FS. Effects of like symptoms in people who use cannabis. Br J Psychiatry ipsapirone and cannabidiol on human experimental anxiety. J 2008;192:306-307. Psychopharmacol 1993;7:82-88. 117. Morgan CJ, Schafer G, Freeman TP, Curran HV. Impact of 101. Martin-Santos R, Crippa JA, Batalla A, et al. Acute effects of a cannabidiol on the acute memory and psychotomimetic effects single, oral dose of d9-tetrahydrocannabinol (THC) and of smoked cannabis: naturalistic study: naturalistic study cannabidiol (CBD) administration in healthy volunteers. Curr [corrected]. Br J Psychiatry 2010;197:285-290. Pharm Design 2012;18:4966-4979. 118. Demirakca T, Sartorius A, Ende G, et al. Diminished gray matter 102. Crippa JA, Zuardi AW, Garrido GE, et al. Effects of in the hippocampus of cannabis users: possible protective effects cannabidiol (CBD) on regional cerebral blood flow. of cannabidiol. Drug Alcohol Depend 2011;114:242-245. Neuropsychopharmacology 2004;29:417-426. 119. Niesink RJ, van Laar MW. Does cannabidiol protect against ad- 103. Bhattacharyya S, Morrison PD, Fusar-Poli P, et al. Opposite ef- verse psychological effects of THC? Front Psychiatry 2013;4:130. fects of delta-9-tetrahydrocannabinol and cannabidiol on human 120. Greer GR, Grob CS, Halberstadt AL. PTSD symptom reports of brain function and psychopathology. Neuropsychopharmacology patients evaluated for the New Mexico Medical Cannabis 2010;35:764-774. Program. J Psychoactive Drugs 2014;46:73-77. 104. Crippa JA, Derenusson GN, Ferrari TB, et al. Neural basis of 121. Passie T, Emrich HM, Karst M, Brandt SD, Halpern JH. anxiolytic effects of cannabidiol (CBD) in generalized social anx- Mitigation of post-traumatic stress symptoms by Cannabis resin: iety disorder: a preliminary report. J Psychopharmacol 2011;25: a review of the clinical and neurobiological evidence. Drug Test 121-130. Anal 2012;4:649-659. Vincent Paolone, M.D. Adult and Adolescent Psychiatry 8166 MarketStreet, Unit B Boardman, OH 44512 Office Phone: (330)758-8528, Office Fax: (330)758-8529 www. board man psychiatrist.co m

December 21, 2020

Dear Medical Board ofthe State of Ohio, lam writingthis letter in supportofadding the diagnosis of Panic Disorderwith Agoraphobia (F40.01) to the approved diagnoses for medical marijuana in Ohio. I am a psychiatrist with almost 30 yearsof experience practicingin the Youngstown area. I became certified for medical marijuana to have another treatment modality available for my patients. I have had many patie nts who experienced therapeutic benefrt f rom marijuana. I have also had manypatients who did not benefit from traditionaltreatments or could not tolerate them. I completely understand the Ohio Medical Board's decision to limit approved d iagnoses for medicalmaruuana. Now that the program hasbeenactive foralmost 3 years,lfeelitis timetoadd Panic Disorde r with Agoraphobia to the list of approved diagnoses. PanicDisorderis a major cost to society. lt isone ofthe major causes for applications forsocial security. I found on line an estimate that panic d isorder and other anxiety d isorde rs cost our society almost 49 billion dollars yearly. Medical marijuana would help limit the use of benzodiazepines, which havethe burden oftolerance and withdrawal. lrespectfullyrequestthattheOhio Medical Board consider adding FzlO.01 to the list of approved diagnoses for med ical marijuana. fulr^ Vincent Paolone, M.D, Ohio Medical License #35066876 Diplomat of the American Board of Psychiatry and Neurology since 1997 Fellow of the American Psychiatric Association since 2014