Defining Risks of Taxane Neuropathy: Insights from Randomized Clinical Trials

Total Page:16

File Type:pdf, Size:1020Kb

Defining Risks of Taxane Neuropathy: Insights from Randomized Clinical Trials Published OnlineFirst July 1, 2013; DOI: 10.1158/1078-0432.CCR-13-0572 Clinical Cancer Review Research Defining Risks of Taxane Neuropathy: Insights from Randomized Clinical Trials David Kudlowitz and Franco Muggia Abstract Sensory neuropathy is a common but difficult to quantify complication encountered during treatment of various cancers with taxane-containing regimens. Docetaxel, paclitaxel, and its nanoparticle albumin- bound formulation have been extensively studied in randomized clinical trials comparing various dose and schedules for the treatment of breast, lung, and ovarian cancers. This review highlights differences in extent of severe neuropathies encountered in such randomized trials and seeks to draw conclusions in terms of known pharmacologic factors that may lead to neuropathy. This basic knowledge provides an essential background for exploring pharmacogenomic differences among patients in relation to their susceptibility of developing severe manifestations. In addition, the differences highlighted may lead to greater insight into drug and basic host factors (such as age, sex, and ethnicity) contributing to axonal injury from taxanes. Clin Cancer Res; 19(17); 1–8. Ó2013 AACR. Introduction dules, whereas the shorter schedule proved safe but had Taxanes have become key drugs in the treatment of more sensory neuropathy. Soon after, docetaxel under- several malignancies as the antitumor activity of paclitaxel went clinical development as Taxotere (under sponsor- and docetaxel was established in the early 1990s. A major ship of Sanofi-Aventis) and became widely used for problem in the clinical use of these drugs, particularly breast and lung cancers (4). The nanoparticle albumin- paclitaxel, has been the development of sensory neurop- bound paclitaxel (nab-paclitaxel or Abraxane) achieved athy. To better define factors resulting in this neurotoxicity, approval for the treatment of breast cancer in 2005. we have reviewed the extent of taxane neuropathy observed Cabazitaxel (5) subsequently obtained U.S. Food and in randomized clinical trials with a particular focus on Drug Administration (FDA) approval for docetaxel- trials where paclitaxel, its nanoparticle albumin-bound refractory prostate cancer and paclitaxel polyglutamate paclitaxel formulation (nab-paclitaxel), and docetaxel are (6) is undergoing registration trials; both are less infor- compared. mative for the purpose of this review. Historically, the contributions of paclitaxel to the treat- The background describes taxane neuropathy and its risk ment of breast, ovarian, and lung cancers became apparent factors such as dose schedule, drug pharmacology, potential in the early 1990s. Bristol-Myers Squibb, once acquiring drug interactions, and preexisting conditions. Clinical trials licensing rights from the National Cancer Institute under in breast, lung, and ovarian cancers are then analyzed to the original generic name Taxol, proceeded to explore (in highlight dose and schedule risk factors for sensory ovarian cancer trials) dose schedule and hypersensitivity neuropathy. issues that had plagued its early development in the 1980s (1). Shorter than the established safe 24-hour Background infusions were explored including a randomized phase Description of taxane neurotoxicity II study with a 2 Â 2 design comparing every 3 weeks 135 Distal symmetrical paresthesias, at first transient and versus 175 mg/m2 doses, and 24- versus 3-hour infusions then constant (with gradual improvement over months to (2, 3). The higher dose was more active in both sche- years without further exposure), are the hallmark of pac- litaxel-induced neuropathy. Other manifestations (such as autonomic and motor changes) are occasionally seen Authors' Affiliation: New York University School of Medicine and Cancer upon coadministration of other neurotoxic drugs or by Institute, New York, New York the presence of preexisting conditions (such as long-stand- Note: Supplementary data for this article are available at Clinical Cancer ing diabetes mellitus). Plantar surfaces at the metatarsal– Research Online (http://clincancerres.aacrjournals.org/). phalangeal junction are often first affected. A frequent Corresponding Author: Franco Muggia, NYU Clinical Cancer Center, Rm finding is distal fingertip paresthesia. Unlike cisplatin- 429, 160 East 34th Street, New York, NY 10016. Phone: 212-263-6485; related neuropathies, taxanes rarely cause loss of distal Fax: 212-263-8210; E-mail: [email protected] deep tendon reflexes (7, 8). Asymmetrical findings usually doi: 10.1158/1078-0432.CCR-13-0572 point to other contributory causes like neurogenic damage Ó2013 American Association for Cancer Research. from discogenic root compression. www.aacrjournals.org OF1 Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 1, 2013; DOI: 10.1158/1078-0432.CCR-13-0572 Kudlowitz and Muggia Paclitaxel was initially paired with cisplatin in the first- in Table 1. Cross-resistance patterns (e.g., with doxorubicin line treatment of ovarian cancer before Gynecologic and vinca alkaloids) reflect, in part, the greater affinity of Oncology Group GOG158 trial showed the noninferiority paclitaxel for P-glycoprotein (P-gp; ref. 16). Moreover, as of the less neurotoxic carboplatin (9). This resulted in CrEL is also a substrate for P-gp, this solubilizing vehicle many women experiencing irreversible life-long neuro- modifies the influx and efflux of paclitaxel, affecting the pathy. When given by itself or with carboplatin, taxane development/reversibility of neuropathy and of myelosup- sensory neuropathy is usually fully reversible unless dosing pression (ref. 17; Fig. 1). However, factors other than P-gp is not modified before incurring in severe neurotoxicity explain the effect of CrEL on paclitaxel PK (18). Differences (10). Retreatment with taxanes is often associated with an in pharmacokinetics and intracellular dynamics may accelerated development of this toxicity. In summary, account for the variable occurrence of neuropathy and neurons conducting pain and touch sensations in the distal myelosuppression, as well as less common effects (e.g., extremities are most vulnerable to taxanes, to other anti- edema, skin rash, and nail changes). Further information tubulin agents, and to platinums (11). Pharmacogenomic on the drug properties of paclitaxel, docetaxel, and nab- studies are seeking to explain an individual susceptibility to paclitaxel is provided in the Supplementary Text (see Drug severe taxane neuropathy (12–15). The current analysis Properties). focuses on specific taxane clinical data on neuropathy from large randomized clinical trials. Methods Neuropathy, myelosuppression, and outcome reported by Drug formulations and pharmacology selected National Cancer Institute (NCI) U.S. cooperative Paclitaxel, docetaxel, and nab-paclitaxel target identical group trials in breast, ovarian, and lung cancers are reviewed. sequences in b-tubulin leading to stabilization of micro- All trials are full publications except for CALBG 40502 (19). tubular dynamics and "bundling." Major pharmacody- In addition, similar data from trials using response evalua- namic differences between these drugs are summarized tion and toxicity criteria adopted by NCI-supported trials are Table 1. Distinguishing pharmacologic features of nab-paclitaxel, paclitaxel, and docetaxel Feature Nab-paclitaxel Paclitaxel Docetaxel Linearity Linear Nonlinear Linear from 75 to 100 mg/m2 Vehicle Albumin CremophorEL and dehydrated Polysorbate 80 and 13% ethanol per ethanol USP (1:1, v/v) 1.5 mL Half-life 21.6 h (17.2 coeff variation 20.5 h (14.6 coeff of variation, for Alpha, beta, gamma 4.5, 38.3, and for 260 mg/m2) 175 mg/m2) 12.2 h Intracellular Albumin leading to tumor Major substrate for P-gp more than Longer intracellular retention time and pharmacodynamic accumulation via SPARC docetaxel (16) higher intracellular concentration in factors (67) target cells (compared with paclitaxel; ref. 68) Substrate for P-gp (69) Clinical delivery 10-min low-volume infusion Infusion rate limited by volume and Rate of delivery volume less also need for special tubing (PDR) constrained than for paclitaxel Issues in oral Not studied Low bioavailability CYP3A4 metabolization (72) bioavailability 1. P-gp transporters found in high Improved by ritonavir (CYP3A4 concentration in the inhibitor and minor P-gp inhibitor; gastrointestinal tract ref. 73) 2. Presystemic metabolism by CYP3A4 and CYP2C8 (70) 3. Improved by GF120918 (71) Intraperitoneal Not studied Ideal dosing in phase I study found to Pharmacologic advantage at 100 therapy be at 60–65 mg/m2 weekly in mg/m2 every 3 wks (76) ovarian cancer (74) Phase 2 trial showed that paclitaxel Lower concentrations than weekly intraperitoneal 60 mg/m2 intraperitoneal paclitaxel, but better with intraperitoneal carboplatin penetrance likely without mycelle every 3 wks had effective systemic formation due to CremophorEL (77) exposure (75) OF2 Clin Cancer Res; 19(17) September 1, 2013 Clinical Cancer Research Downloaded from clincancerres.aacrjournals.org on October 1, 2021. © 2013 American Association for Cancer Research. Published OnlineFirst July 1, 2013; DOI: 10.1158/1078-0432.CCR-13-0572 Defining Risks of Taxane Neuropathy similar peripheral neuropathy when compared with con- CrEL CrEL current therapy (27). In 2005, the registration trial for nab-paclitaxel uPAC CrEL compared nab-paclitaxel 260 mg/m2 with paclitaxel N CrEL CrEL 175 mg/m2 (each every 3 weeks).
Recommended publications
  • Pemetrexed-Lilly-Epar-Product-Information En.Pdf
    ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS 1 1. NAME OF THE MEDICINAL PRODUCT Pemetrexed Lilly 100 mg powder for concentrate for solution for infusion Pemetrexed Lilly 500 mg powder for concentrate for solution for infusion 2. QUALITATIVE AND QUANTITATIVE COMPOSITION Pemetrexed Lilly 100 mg powder for concentrate for solution for infusion Each vial contains 100 mg of pemetrexed (as pemetrexed disodium). Excipient with known effect Each vial contains approximately 11 mg sodium. Pemetrexed Lilly 500 mg powder for concentrate for solution for infusion Each vial contains 500 mg of pemetrexed (as pemetrexed disodium). Excipient with known effect Each vial contains approximately 54 mg sodium. After reconstitution (see section 6.6), each vial contains 25 mg/mL of pemetrexed. For the full list of excipients, see section 6.1. 3. PHARMACEUTICAL FORM Powder for concentrate for solution for infusion. White to either light yellow or green-yellow lyophilised powder. 4. CLINICAL PARTICULARS 4.1 Therapeutic indications Malignant pleural mesothelioma Pemetrexed Lilly in combination with cisplatin is indicated for the treatment of chemotherapy naïve patients with unresectable malignant pleural mesothelioma. Non-small cell lung cancer Pemetrexed Lilly in combination with cisplatin is indicated for the first line treatment of patients with locally advanced or metastatic non-small cell lung cancer other than predominantly squamous cell histology (see section 5.1). Pemetrexed Lilly is indicated as monotherapy for the maintenance treatment of locally advanced or metastatic non-small cell lung cancer other than predominantly squamous cell histology in patients whose disease has not progressed immediately following platinum-based chemotherapy (see section 5.1).
    [Show full text]
  • Pemetrexed (Alimta) Is Covered Pemetrexed (Alimta®) Is Considered Medically Necessary for the Treatment of Patients With
    Corporate Medical Policy ® Pemetrexed (Alimta ) File Name: pemetrexed_alimta Origination: 8/2016 Last CAP Review: 4/2020 Next CAP Review: 4/2021 Last Review: 4/2020 Description of Procedure or Service ® Pemetrexed (Alimta ) is a folate analog metabolic inhibitor indicated for non-squamous non-small cell lung cancer, mesothelioma, urothelial carcinoma, epithelial ovarian cancer and thymic carcinoma. Alimta is indicated in combination with cisplatin therapy for the initial treatment of patients with locally advanced or metastatic non-squamous non-small cell lung cancer, and in combination with pembrolizumab and platinum chemotherapy for the initial treatment of patients with metastatic non- squamous non-small cell lung cancer and with no EGFR or ALK genomic tumor aberrations. Alimta is indicated for the maintenance treatment of patients with locally advanced or metastatic non-squamous non-small cell lung cancer whose disease has not progressed after four cycles of platinum-based first- line chemotherapy. Alimta is indicated as a single agent for the treatment of patients with recurrent, metastatic non- squamous non-small cell lung cancer after prior chemotherapy. Alimta in combination with cisplatin is indicated for the treatment of patients with malignant pleural mesothelioma whose disease is unresectable or who are otherwise not candidates for curative surgery. ***Note: This Medical Policy is complex and technical. For questions concerning the technical language and/or specific clinical indications for its use, please consult your physician. Policy ® BCBSNC will provide coverage for Pemetrexed (Alimta ) when it is determined to be medically necessary because the medical criteria and guidelines noted below are met. Benefits Application This medical policy relates only to the services or supplies described herein.
    [Show full text]
  • Original Article Pemetrexed and Cyclophosphamide Combination Therapy for the Treatment of Non-Small Cell Lung Cancer
    Int J Clin Exp Pathol 2015;8(11):14693-14700 www.ijcep.com /ISSN:1936-2625/IJCEP0012945 Original Article Pemetrexed and cyclophosphamide combination therapy for the treatment of non-small cell lung cancer Dong Li1, Song He2 1Department of Cardiothoracic Surgery, Central Hospital of Zibo, Zibo 250012, Shandong, China; 2Maanshan Center for Clinical Laboratory, Maanshan Municipal Hospital Group, Maanshan, Anhui, China Received July 15, 2015; Accepted October 13, 2015; Epub November 1, 2015; Published November 15, 2015 Abstract: Lung cancer is the leading cause of cancer-related mortality. This study was undertaken to investigate the efficacy and safety of adding regulatory T cell inhibitor cyclophosphamide to pemetrexed therapy for the second-line treatment of NSCLC with wild-type epidermal growth factor receptor (EGFR). A total of 70 patients were screened between March 2011 and December 2013, out of which 62 patients were enrolled in the study. Patients were randomized to receive 500 mg/m2 pemetrexed in combination with 20 mg/kg cyclophosphamide in a 21 day cycle (n=30) or 500 mg/m2 pemetrexed (n=32), and followed up for 30 months. Disease progression was observed in 23 patients in the pemetrexed plus cyclophosphamide arm and 27 patients in the pemetrexed monotherapy arm. Median progression-free survival was 3.6 months (95% confidence interval [CI], 1.3 to 5.9 months) in the peme- trexed plus cyclophosphamide arm and 2.2 months (95% CI, 1.3 to 3.1 months) in the pemetrexed monotherapy arm. The 6-month PFS rates were 22% (95% CI, 10 to 34) and 14.5% (95% CI, 6 to 23) in the pemetrexed plus cyclophosphamide arm and pemetrexed monotherapy arm, respectively.
    [Show full text]
  • A Preclinical Evaluation of Pemetrexed and Irinotecan Combination As Second-Line Chemotherapy in Pancreatic Cancer
    British Journal of Cancer (2007) 96, 1358 – 1367 & 2007 Cancer Research UK All rights reserved 0007 – 0920/07 $30.00 www.bjcancer.com A preclinical evaluation of pemetrexed and irinotecan combination as second-line chemotherapy in pancreatic cancer A Mercalli1, V Sordi1, R Formicola1, M Dandrea2, S Beghelli2, A Scarpa2, V Di Carlo1, M Reni3,4 and L Piemonti*,1,4 1 2 Laboratory of Experimental Surgery, San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy; Section of Anatomic Pathology, Department 3 of Pathology, University of Verona, Strada Le Grazie 8, Verona 37134, Italy; Department of Oncology, San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy Translational Therapeutics Gemcitabine (GEM)-based chemotherapy is regarded as the standard treatment of pancreatic adenocarcinoma, but yields a very limited disease control. Very few studies have investigated salvage chemotherapy after failure of GEM or GEM-containing chemotherapy and preclinical studies attempting to widen the therapeutic armamentarium, not including GEM, are warranted. MIA PaCa2, CFPAC-1 and Capan-1 pancreatic cancer cell lines were treated with GEM, fluouracil (5-FU), docetaxel (DCT), oxaliplatin (OXP), irinotecan (CPT-11), pemetrexed (PMX) and raltitrexed (RTX) as single agent. Pemetrexed, inducing apoptosis with IC50s under the Cmax in the three lines tested, appeared the most effective drug as single agent. Based on these results, schedule- and concentration-dependent drug interactions (assessed using the combination index) of PMX/GEM, PMX/DCT and PMX–CPT-11 were evaluated. The combinatory study clearly indicated the PMX and CPT-11 combination as the most active against pancreatic cancer. To confirm the efficacy of PMX–CPT-11 combination, we extended the study to a panel of 10 pancreatic cancer cell lines using clinically relevant concentrations (PMX 10 mM; CPT-11 1 mm).
    [Show full text]
  • Cancer Drug Pharmacology Table
    CANCER DRUG PHARMACOLOGY TABLE Cytotoxic Chemotherapy Drugs are classified according to the BC Cancer Drug Manual Monographs, unless otherwise specified (see asterisks). Subclassifications are in brackets where applicable. Alkylating Agents have reactive groups (usually alkyl) that attach to Antimetabolites are structural analogues of naturally occurring molecules DNA or RNA, leading to interruption in synthesis of DNA, RNA, or required for DNA and RNA synthesis. When substituted for the natural body proteins. substances, they disrupt DNA and RNA synthesis. bendamustine (nitrogen mustard) azacitidine (pyrimidine analogue) busulfan (alkyl sulfonate) capecitabine (pyrimidine analogue) carboplatin (platinum) cladribine (adenosine analogue) carmustine (nitrosurea) cytarabine (pyrimidine analogue) chlorambucil (nitrogen mustard) fludarabine (purine analogue) cisplatin (platinum) fluorouracil (pyrimidine analogue) cyclophosphamide (nitrogen mustard) gemcitabine (pyrimidine analogue) dacarbazine (triazine) mercaptopurine (purine analogue) estramustine (nitrogen mustard with 17-beta-estradiol) methotrexate (folate analogue) hydroxyurea pralatrexate (folate analogue) ifosfamide (nitrogen mustard) pemetrexed (folate analogue) lomustine (nitrosurea) pentostatin (purine analogue) mechlorethamine (nitrogen mustard) raltitrexed (folate analogue) melphalan (nitrogen mustard) thioguanine (purine analogue) oxaliplatin (platinum) trifluridine-tipiracil (pyrimidine analogue/thymidine phosphorylase procarbazine (triazine) inhibitor)
    [Show full text]
  • Re-Visiting Hypersensitivity Reactions to Taxanes: a Comprehensive Review
    Clinic Rev Allerg Immunol DOI 10.1007/s12016-014-8416-0 Re-visiting Hypersensitivity Reactions to Taxanes: A Comprehensive Review Matthieu Picard & Mariana C. Castells # Springer Science+Business Media New York 2014 Abstract Taxanes (a class of chemotherapeutic agents) Keywords Taxane . Paclitaxel . Taxol . Docetaxel . are an important cause of hypersensitivity reactions Taxotere . Nab-paclitaxel . Abraxane . Cabazitaxel . (HSRs) in cancer patients. During the last decade, the Chemotherapy . Hypersensitivity . Allergy . Skin test . development of rapid drug desensitization has been key Desensitization . Challenge . Diagnosis . Review . IgE . to allow patients with HSRs to taxanes to be safely re- Complement . Mechanism treated although the mechanisms of these HSRs are not fully understood. Earlier studies suggested that solvents, such as Cremophor EL used to solubilize paclitaxel, Introduction were responsible for HSRs through complement activa- tion, but recent findings have raised the possibility that Hypersensitivity reactions (HSRs) to chemotherapy are in- some of these HSRs are IgE-mediated. Taxane skin creasingly common and represent an important impediment testing, which identifies patients with an IgE-mediated to the care of cancer patients as they may entail serious sensitivity, appears as a promising diagnostic and risk consequences and prevent patients from being treated with stratification tool in the management of patients with the most efficacious agent against their cancer [1]. During the HSRs to taxanes. The management of patients following last decade, different groups have developed rapid drug de- a HSR involves risk stratification and re-exposure could sensitization (RDD) protocols that allow the safe re- be performed either through rapid drug desensitization introduction of a chemotherapeutic agent to which a patient or graded challenge based on the severity of the initial is allergic, and their use have recently been endorsed by the HSR and the skin test result.
    [Show full text]
  • BC Cancer Benefit Drug List September 2021
    Page 1 of 65 BC Cancer Benefit Drug List September 2021 DEFINITIONS Class I Reimbursed for active cancer or approved treatment or approved indication only. Reimbursed for approved indications only. Completion of the BC Cancer Compassionate Access Program Application (formerly Undesignated Indication Form) is necessary to Restricted Funding (R) provide the appropriate clinical information for each patient. NOTES 1. BC Cancer will reimburse, to the Communities Oncology Network hospital pharmacy, the actual acquisition cost of a Benefit Drug, up to the maximum price as determined by BC Cancer, based on the current brand and contract price. Please contact the OSCAR Hotline at 1-888-355-0355 if more information is required. 2. Not Otherwise Specified (NOS) code only applicable to Class I drugs where indicated. 3. Intrahepatic use of chemotherapy drugs is not reimbursable unless specified. 4. For queries regarding other indications not specified, please contact the BC Cancer Compassionate Access Program Office at 604.877.6000 x 6277 or [email protected] DOSAGE TUMOUR PROTOCOL DRUG APPROVED INDICATIONS CLASS NOTES FORM SITE CODES Therapy for Metastatic Castration-Sensitive Prostate Cancer using abiraterone tablet Genitourinary UGUMCSPABI* R Abiraterone and Prednisone Palliative Therapy for Metastatic Castration Resistant Prostate Cancer abiraterone tablet Genitourinary UGUPABI R Using Abiraterone and prednisone acitretin capsule Lymphoma reversal of early dysplastic and neoplastic stem changes LYNOS I first-line treatment of epidermal
    [Show full text]
  • Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics
    Review Current Advances of Nitric Oxide in Cancer and Anticancer Therapeutics Joel Mintz 1,†, Anastasia Vedenko 2,†, Omar Rosete 3 , Khushi Shah 4, Gabriella Goldstein 5 , Joshua M. Hare 2,6,7 , Ranjith Ramasamy 3,6,* and Himanshu Arora 2,3,6,* 1 Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL 33328, USA; [email protected] 2 John P Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; [email protected] (A.V.); [email protected] (J.M.H.) 3 Department of Urology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; [email protected] 4 College of Arts and Sciences, University of Miami, Miami, FL 33146, USA; [email protected] 5 College of Health Professions and Sciences, University of Central Florida, Orlando, FL 32816, USA; [email protected] 6 The Interdisciplinary Stem Cell Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA 7 Department of Medicine, Cardiology Division, Miller School of Medicine, University of Miami, Miami, FL 33136, USA * Correspondence: [email protected] (R.R.); [email protected] (H.A.) † These authors contributed equally to this work. Abstract: Nitric oxide (NO) is a short-lived, ubiquitous signaling molecule that affects numerous critical functions in the body. There are markedly conflicting findings in the literature regarding the bimodal effects of NO in carcinogenesis and tumor progression, which has important consequences for treatment. Several preclinical and clinical studies have suggested that both pro- and antitumori- Citation: Mintz, J.; Vedenko, A.; genic effects of NO depend on multiple aspects, including, but not limited to, tissue of generation, the Rosete, O.; Shah, K.; Goldstein, G.; level of production, the oxidative/reductive (redox) environment in which this radical is generated, Hare, J.M; Ramasamy, R.; Arora, H.
    [Show full text]
  • Arsenic Trioxide Targets MTHFD1 and SUMO-Dependent Nuclear De Novo Thymidylate Biosynthesis
    Arsenic trioxide targets MTHFD1 and SUMO-dependent PNAS PLUS nuclear de novo thymidylate biosynthesis Elena Kamyninaa, Erica R. Lachenauera,b, Aislyn C. DiRisioa, Rebecca P. Liebenthala, Martha S. Fielda, and Patrick J. Stovera,b,c,1 aDivision of Nutritional Sciences, Cornell University, Ithaca, NY 14853; bGraduate Field of Biology and Biomedical Sciences, Cornell University, Ithaca, NY 14853; and cGraduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853 Contributed by Patrick J. Stover, February 12, 2017 (sent for review December 1, 2016; reviewed by I. David Goldman and Anne Parle-McDermott) Arsenic exposure increases risk for cancers and is teratogenic in levels. Decreased rates of de novo dTMP synthesis can be caused animal models. Here we demonstrate that small ubiquitin-like by the action of chemotherapeutic drugs (19), through inborn modifier (SUMO)- and folate-dependent nuclear de novo thymidylate errors of folate transport and metabolism (15, 18, 20, 21), by (dTMP) biosynthesis is a sensitive target of arsenic trioxide (As2O3), inhibiting translocation of the dTMP synthesis pathway enzymes leading to uracil misincorporation into DNA and genome instability. into the nucleus (2) and by dietary folate deficiency (22, 23). Im- Methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) and serine paired dTMP synthesis leads to genome instability through well- hydroxymethyltransferase (SHMT) generate 5,10-methylenetetrahy- characterized mechanisms associated with uracil misincorporation drofolate for de novo dTMP biosynthesis and translocate to the nu- into nuclear DNA and subsequent futile cycles of DNA repair (24, cleus during S-phase, where they form a multienzyme complex with 25). Nuclear DNA is surveyed for the presence of uracil by a thymidylate synthase (TYMS) and dihydrofolate reductase (DHFR), as family of uracil glycosylases including: uracil N-glycolase (UNG), well as the components of the DNA replication machinery.
    [Show full text]
  • Filed by Cell Therapeutics, Inc. Pursuant to Rule 425 Under The
    Filed by Cell Therapeutics, Inc. Pursuant to Rule 425 under the Securities Act of 1933 And deemed filed pursuant Rule 14a-12 Of the Securities Exchange Act of 1934 Subject Company: Cell Therapeutics, Inc. Commission File No.: 001-12465 The following is a transcript of a presentation given by Cell Therapeutics, Inc. at its annual meeting of shareholders, held on June 20, 2003. Moderator: Jim Bianco Operator: Good day everyone and welcome to the Cell Therapeutics annual shareholder meeting. As a reminder, this call is being recorded. We’ll soon be going live to Seattle where the call will begin shortly. Please stand by. Jim Bianco: Welcome. My name is Jim Bianco. I’m the President and CEO of Cell Therapeutics. Welcome to our 2003 shareholders meeting. Our business meeting agenda today is to approve the minutes, elect the directors, and approve the equity incentive plan as well as the amendment to the employees stock purchase plan, and ratify the selection of E&Y as independent auditors. Mike Kennedy, the Secretary of the company, will act as secretary of this meeting, and George Pabst has been appointed inspector of elections to examine and count proxies and ballots. At the conclusion of the business portion of today’s meeting, members of management will present highlights from the past year and outline some of our future milestones and objectives for the next 12 to 18 months. At this time, I’d like to call the meeting to order. Let me begin by introducing our directors who are present today and let me start by saying that Dr.
    [Show full text]
  • Pharmacogenomic Biomarkers in Docetaxel Treatment of Prostate Cancer: from Discovery to Implementation
    G C A T T A C G G C A T genes Review Pharmacogenomic Biomarkers in Docetaxel Treatment of Prostate Cancer: From Discovery to Implementation Reka Varnai 1,2, Leena M. Koskinen 3, Laura E. Mäntylä 3, Istvan Szabo 4,5, Liesel M. FitzGerald 6 and Csilla Sipeky 3,* 1 Department of Primary Health Care, University of Pécs, Rákóczi u 2, H-7623 Pécs, Hungary 2 Faculty of Health Sciences, Doctoral School of Health Sciences, University of Pécs, Vörösmarty u 4, H-7621 Pécs, Hungary 3 Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland 4 Institute of Sport Sciences and Physical Education, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary 5 Faculty of Sciences, Doctoral School of Biology and Sportbiology, University of Pécs, Ifjúság útja 6, H-7624 Pécs, Hungary 6 Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia * Correspondence: csilla.sipeky@utu.fi Received: 17 June 2019; Accepted: 5 August 2019; Published: 8 August 2019 Abstract: Prostate cancer is the fifth leading cause of male cancer death worldwide. Although docetaxel chemotherapy has been used for more than fifteen years to treat metastatic castration resistant prostate cancer, the high inter-individual variability of treatment efficacy and toxicity is still not well understood. Since prostate cancer has a high heritability, inherited biomarkers of the genomic signature may be appropriate tools to guide treatment. In this review, we provide an extensive overview and discuss the current state of the art of pharmacogenomic biomarkers modulating docetaxel treatment of prostate cancer. This includes (1) research studies with a focus on germline genomic biomarkers, (2) clinical trials including a range of genetic signatures, and (3) their implementation in treatment guidelines.
    [Show full text]
  • Exosomes and Breast Cancer Drug Resistance Xingli Dong1,2, Xupeng Bai 2,3,Jieni2,3,Haozhang 4,Weiduan5, Peter Graham2,3 Andyongli 2,3,6
    Dong et al. Cell Death and Disease (2020) 11:987 https://doi.org/10.1038/s41419-020-03189-z Cell Death & Disease REVIEW ARTICLE Open Access Exosomes and breast cancer drug resistance Xingli Dong1,2, Xupeng Bai 2,3,JieNi2,3,HaoZhang 4,WeiDuan5, Peter Graham2,3 andYongLi 2,3,6 Abstract Drug resistance is a daunting challenge in the treatment of breast cancer (BC). Exosomes, as intercellular communicative vectors in the tumor microenvironment, play an important role in BC progression. With the in-depth understanding of tumor heterogeneity, an emerging role of exosomes in drug resistance has attracted extensive attention. The functional proteins or non-coding RNAs contained in exosomes secreted from tumor and stromal cells mediate drug resistance by regulating drug efflux and metabolism, pro-survival signaling, epithelial–mesenchymal transition, stem-like property, and tumor microenvironmental remodeling. In this review, we summarize the underlying associations between exosomes and drug resistance of BC and discuss the unique biogenesis of exosomes, the change of exosome cargo, and the pattern of release by BC cells in response to drug treatment. Moreover, we propose exosome as a candidate biomarker in predicting and monitoring the therapeutic drug response of BC and as a potential target or carrier to reverse the drug resistance of BC. ● Facts Tumor-derived exosomes mediate enhanced EMT and stem-like property of drug-resistant BC. ● ● Tumor-derived exosomes mediate the chemoresistance TME-derived exosomes mediate the tumor of BC by reducing the intracellular accumulation of microenvironmental remodeling that favors the drug resistance of BC. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; chemotherapeutic drugs and delivering functional ● cargos that activate pro-survival signaling and The exosome is proposed as a candidate biomarker in unchecked cell cycle progression.
    [Show full text]