New Antibiotic Approvals 2017-2019 Joshua Garcia, Pharmd, BCPS Assistant Professor, Marshall B

Total Page:16

File Type:pdf, Size:1020Kb

New Antibiotic Approvals 2017-2019 Joshua Garcia, Pharmd, BCPS Assistant Professor, Marshall B New Antibiotic Approvals 2017-2019 Joshua Garcia, PharmD, BCPS Assistant Professor, Marshall B. Ketchum University May 12th 2020 Conflicts of Interest No conflicts of interests to disclose Objectives Identify indications for new, recently approved antimicrobials Recognize pros and cons of using newly approved antimicrobials in a clinical situation Identify major adverse effect, monitoring parameters, and clinical pearls of newly approved antimicrobials Antimicrobial Resistance Problem Updated numbers: 2,800,000 illnesses and 35,000 deaths IDSA 10 x20 initiative CDC Antimicrobial Resistance Reports, 2013 and 2019 GAIN Act – 2012 Limited Population Antibacterial Drug (LPAD) pathway 2016 Antibacterial Resistance Leadership Group of the National Institutes of Health (NIH) Combating Antibiotic-Resistant Bacteria Biopharmaceutical Accelerator (CARB-X) The Antibiotic Resistance Problem: The General Idea Selective pressure (antibiotics) = Rise of resistant organisms Wild-type bacteria Bacteria with resistance gene The Antibiotic Resistance Problem: The Macroscale Selective pressures Inappropriate antibiotics In the US: CDC estimated 30-50% (47 million courses) of all prescribed antibiotics are inappropriate / unnecessary Increasing use of broad spectrum antibiotics Antibiotic use in livestock and environment Dearth of new antimicrobials in development 1. Dellit TH, et al. Clin Infect Dis. 2007 Jan 15;44(2):159-77. 2. Fridkin SK, et al. MMWR Morb Mortal Wkly Rep. 2014;63(9):194-200 3. Fleming-Dutra KE, et al. JAMA. 2016 May 3;315(17):1864-73. doi: 10.1001/jama.2016.4151. 4. Woolhouse M, et al. Philos Trans R Soc Lond B Biol Sci. 2015 Jun 5; 370(1670): 20140083. doi: 10.1098/rstb.2014.0083 The Antibiotic Resistance Problem: The Microscale Antibiotic Inactivation Active Efflux Ex. Beta-lacatamase production Ex. Tet(A) efflux pumps for tetracyclines Porin Channel Target Alteration Degradation Ex. OprD porin mutations in Pseudomonas aeruginosa Ex. mecA gene in MRSA Antibiotic Approval Timeline Antibiotic Deployment Antibiotic Resistance Clatworthy et al. Nat Chem Biol. 2007 Sep;3(9):541-8 Antibiotic Approval Timeline: Updated Antibiotic Deployment Dalbavancin Oritavancin Tedizolid Avycaz Zerbaxa 2010 2015 2020 Antibiotic Resistance Clatworthy et al. Nat Chem Biol. 2007 Sep;3(9):541-8 New Antibiotic Approvals: 2017-2019 Brand Name Generic Name Approval Date BaxdelaTM Delafloxacin 6/2017 VabormereTM Meropenem/vaborbactam 8/2017 ZemdriTM Plazomicin 7/2018 XeravaTM Eravacycline 8/2018 NuzyraTM Omadacycline 10/2018 RecarbrioTM Imipenem/cilastatin/relebactam 7/2019 XenletaTM Lefamulin 8/2019 Fetroja® Cefiderocol 11/2019 New Antibiotic Approvals by Intended Use / Indications Acute Bacterial Skin and Skin Structure Infections (ABSSSI) Delafloxacin Resistant Gram Negative Bacilli Omadacycline Meropenem/vaborbactam Plazomicin Eravacycline Community Acquired Pneumonia (CAP) Imipenem/cilastatin/relebactam Omadacycline Cefiderocol Lefamulin Delafloxacin (BaxdelaTM) Delafloxacin (BaxdelaTM) Indication: Acute bacterial skin and skin structure infections (ABSSSI) Spectrum of activity: Activity against S. pyogenes, S. pneumoniae, enteric gram negative rods, M. cattharalis, H. influenzae, B. fragilis, atypical organisms, S. aureus (including MRSA), and P. aeruginosa Mechanism of action: Enhanced inhibition of DNA gyrase and topoisomerase IV due to addition of chlorine at C-8 and removal of basic group at C-7 Dosing: PO: 450mg twice daily IV: 300mg every 12 hours Renal impairment: PO: No adjustment IV: eGFR 15-29 – 200mg every 12 hours Not recommended in ESRD 1. Baxdela [package insert]. 2017 2. Mogle BT, et al. Journal of Antimicrobial Chemotherapy. 2018;73(6):1439-1451. doi:10.1093/jac/dkx543 Delafloxacin (BaxdelaTM) Metabolism Negligible Elimination IV: 65% urine, 28% feces PO: 50% urine, 48% feces Half-life 4.2 – 8.5 hours ADRs: Common: Nausea, diarrhea, vomiting, headache, transaminase elevations FQ Black boxed warnings: Tendon rupture, peripheral neuropathy, CNS effects, exacerbation of myasthenia gravis, hypoglycemia, AMS, aortic dissection Drug Interactions : Oral chelation with cations 1. Baxdela [package insert]. 2017 2. Mogle BT, et al. Journal of Antimicrobial Chemotherapy. 2018;73(6):1439-1451. doi:10.1093/jac/dkx543 Delafloxacin Clinical Trials Trial 1 (IV only) Trial 2 (IV to PO) Design Phase 3, multi-center, double-blind, randomized, non-inferiority Intervention • Delafloxacin 300mg IV BID • Delafloxacin 300mg IV BID for 3 • Vancomycin 15mg/kg + days, then switch to 450mg PO BID aztreonam 2g IV BID • Vancomycin 15mg/kg + aztreonam 2g IV BID Primary Efficacy Objective response at 48-72 hrs / clinical cure Outcome Exclusions Previous ABSSI antibiotic Pregnant or lactating Deeper tissue infection (bone, joint, etc.) or shock Immunocompromised ESRD or liver dysfunction > 10% body burns 1. Pullman J, et al. J Antimicrob Chemother. 2017;72(12):3471-3480. doi:10.1093/jac/dkx329 2. O’Riordan W, et al. Clin Infect Dis. 2018;67(5):657-666. doi:10.1093/cid/ciy165 Trial 1 Delafloxacin Efficacy Results Trial 2 1. Pullman J, et al. J Antimicrob Chemother. 2017;72(12):3471-3480. doi:10.1093/jac/dkx329 2. O’Riordan W, et al. Clin Infect Dis. 2018;67(5):657-666. doi:10.1093/cid/ciy165 S. Aureus Response Comparison Trial 1 Trial 2 1. Pullman J, et al. J Antimicrob Chemother. 2017;72(12):3471-3480. doi:10.1093/jac/dkx329 2. O’Riordan W, et al. Clin Infect Dis. 2018;67(5):657-666. doi:10.1093/cid/ciy165 3. McCurdy S, Lawrence L, Quintas M, et al. Antimicrob Agents Chemother. 2017;61(9):e00772-17, e00772-17. doi:10.1128/AAC.00772-17 Trial 1 Delafloxacin Safety Results Trial 2 • No FQ related C. diff infections, tendon • 1 FQ related C. diff infection (delafloxacin), rupture, or peripheral neuropathy, tendon rupture, or peripheral neuropathy photosensitivity, or Qt prolongation • No tendon rupture, or peripheral neuropathy, • Equal delafloxacin hypo(hyper)glycemia photosensitivity, Qt prolongation or and vancomycin/aztreonam dysglycemia in either group 1. Pullman J, et al. J Antimicrob Chemother. 2017;72(12):3471-3480. doi:10.1093/jac/dkx329 2. O’Riordan W, et al. Clin Infect Dis. 2018;67(5):657-666. doi:10.1093/cid/ciy165 A Deeper Look into Safety Photosensitivity Qtc Prolongation 1. Litwin JS, et al. Antimicrob Agents Chemother. 2015;59(6):3469-3473. doi:10.1128/AAC.04813-14 2. Dawe RS, et al. Photochem Photobiol Sci. 2018;17(6):773-780. doi:10.1039/C8PP00019K Delafloxacin Takeaways / Potential Places in Therapy Takeaways: Unique coverage of both Pseudomonas aeruginosa AND MRSA Less likely to use as routine ABSSSI No labeled warnings/adverse affects of Qt prolongation or photosensitivity Potential Places in Therapy: Step down oral therapy for polymicrobial diabetic foot infections Step down oral therapy for bloodstream infections Community Acquired Bacterial Pneumonia (DEFINE-CABP) Omadacycline (NuzyraTM) Omadacycline (NuzyraTM) Indication: Community acquired bacterial pneumonia (CABP) and acute bacterial skin and skin structure infections (ABSSSI) Spectrum of activity: Streptococcus, E. faecalis, E. faecium, S. aureus (MSSA and MRSA), B. fragilis, atypical organisms, M. cattharalis, H. influenzae Potential coverage of vancomycin-resistant enterococci (VRE), Acinetobacter spp., and ESBL (extended-spectrum beta-lacatamase) producing organisms Mechanism of action: Aminomethyl group modification at C-9 allows for sustained protein synthesis inhibition in the presence of tetracycline efflux pumps Dosing: IV Load PO Load 1. Nuzyra [package insert]. 2018 2. Barber KE, et al. Pharmacotherapy. 2018;38(12):1194-1204. doi:10.1002/phar.2185 Omadacycline (NuzyraTM) Metabolism Negligible Elimination 27% urine, 73% feces Half-life ~16 hrs at steady state ADRs: Nausea, vomiting, diarrhea, transaminase elevations, hypertension Drug Interactions: Anticoagulants: Depression of prothrombin activity may require dose decrease of anticoagulants Oral chelation with cations 1. Nuzyra [package insert]. 2018 2. Barber KE, et al. Pharmacotherapy. 2018;38(12):1194-1204. doi:10.1002/phar.2185 Omadacycline Clinical Trials OPTIC Trial (PNA) OASIS-1 (ABSSSI, IV-PO) OASIS-2 (ABSSSI, PO Only) Design Phase 3, multi-center, double-blind, randomized, non-inferiority Intervention • Omadacycline 100mg IV BID x2 • Omadacycline 100mg IV BID x2 • Omadacycline 450mg PO doses, then 100mg IV daily doses, then 100mg IV daily BID for 2 days, then 300mg (optional transition to 300mg PO (optional transition to 300mg PO daily after 3 days) PO after 3 days) • Linezolid 600mg PO BID • Moxifloxacin 400mg IV daily • Linezolid 600mg IV BID (optional switch to 400mg PO (optional transition to 600mg after 3 days) PO BID after 3 days) Primary Efficacy • Early clinical response (72 – 120 Early clinical response (20% reduction in lesion size at 48 – 72 hours Outcome hours after first dose) after first dose) Exclusions • Previous antibiotic in last 72 hrs Previous antibiotic in last 72 hrs • Hospital acquired PNA Skin lesions < 75cm2 • PSI class V patients Anticipated treatment > 14 days • Renal or hepatic insufficiency Chronic wounds, bites, or diabetic foot infections • Immunocompromised Renal or hepatic insufficiency Immunocompromised 1. Stets R, et al. N Engl J Med. 2019;380(6):517-527. doi:10.1056/NEJMoa1800201 2. O’Riordan W, et al. N Engl J Med. 2019;380(6):528-538. doi:10.1056/NEJMoa1800170 3. O’Riordan W, et al. Lancet Infect Dis. 2019;19(10):1080-1090. doi:10.1016/S1473-3099(19)30275-0 Omadacycline Efficacy Results OPTIC Moxifloxacin better OASIS-1 OASIS-2
Recommended publications
  • IDSA Guideline Review
    1 Infectious Diseases Society of America Antimicrobial Resistant Treatment Guidance: Gram- Negative Bacterial Infections A Focus on Extended-Spectrum β-lactamase Producing Enterobacterales (ESBL-E), Carbapenem- Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Difficult-to-Treat Resistance (DTR- P. aeruginosa) Guideline Summary by Kendra Suder, PharmD Candidate General Recommendations Considerations for choosing empiric therapy: o Local susceptibility patterns/ antibiogram o Patient’s previous organisms isolated & susceptibility data in the last 6 months o Antibiotic exposure in the last 30 days What if a patient was empirically placed on an antibiotic regimen that is now considered inactive against the organism based on susceptibility data? o If an inactive agent was started empirically for cystitis and patient improves, no change in antibiotic or extension of therapy is necessary o However, for other types of infections, guidelines recommend a change to an active regimen for a full treatment course (dated from the start of active therapy) Extended-Spectrum β-Lactamase-Producing Enterobacterales (ESBL-E) Extended-spectrum β-lactamase o Enzymes that inactivate most penicillins, cephalosporins, and aztreonam o Most commonly produced by Escherichia coli, Klebsiella pneumoniae, Klebsiella oxytoca, and Proteus mirabilis (guideline recommendations refer to ESBL-E infections caused by these organisms) o Most common type in the US is the CTX-M, especially CTX-M-15 In institutions that do not perform ESBL testing, a lack of
    [Show full text]
  • Plazomicin Sulfate ASHP INJECTABLE DRUG INFORMATION
    1302 PLAZOMICIN SULFATE ASHP INJECTABLE DRUG INFORMATION Plazomicin Sulfate AHFS 8:12.02 Products Stability Plazomicin sulfate is available in a concentration equivalent Intact vials of plazomicin sulfate should be stored under refrig- to plazomicin base 50 mg/mL in 10-mL single-dose (preserva- eration at 2 to 8°C.3431 Plazomicin sulfate injection is a clear, tive-free) vials.3431 Each vial also contains sodium hydroxide colorless to yellow solution.3431 The manufacturer states that for pH adjustment and water for injection.3431 The appropriate the solution may become yellow, but that this change does not dose of plazomicin solution should be diluted in sodium chloride indicate a decrease in potency.3431 0.9% or Ringer’s injection, lactated to achieve a final volume of The manufacturer states that a solution of plazomicin diluted 3431 50 mL. for infusion in sodium chloride 0.9% or Ringer’s injection, pH lactated to concentrations of 2.5 to 45 mg/mL is stable for up to 24 hours at room temperature.3431 Adjusted to 6.5.3431 Trade Name(s) Zemdri Administration Plazomicin sulfate is administered by intravenous infusion over 30 minutes after dilution in sodium chloride 0.9% or Ringer’s injection, lactated.3431 Compatibility Information Solution Compatibility Plazomicin sulfate Test Soln Name Mfr Mfr Conc/L or % Remarks Ref C/I Ringer’s injection, lactated ACH 2.5 to 45 g Stable for 24 hr at room temperature 3431 C Sodium chloride 0.9% ACH 2.5 to 45 g Stable for 24 hr at room temperature 3431 C Y-Site Injection Compatibility (1:1 Mixture) Plazomicin
    [Show full text]
  • The Pharmacodynamics of Plazomicin and Amikacin Studied in an in Vitro
    P203 The pharmacodynamics of plazomicin and amikacin studied in an in vitro pharmacokinetic model Karen E Bowker 1, Alan R Noel 1, Marie A Attwood 1, Sharon G Tomaselli 1, Alasdair P MacGowan 1, Kevin Krause 2, Eileen Kim 2 BCARE, Department of Microbiology, North Bristol NHS Trust, Bristol, UK. 1 Achaogen Inc, South San Francisco, California, USA 2 [email protected] ASM MICROBE, New Orleans, 1-5th June, 2017 Introduction Results The AUC/MIC targets for amikacin administered 12hrly against E.coli (n=3) Aminoglycoside antibiotics have been a mainstay of antimicrobial Table 3: Individual and mean AUC/MIC targets for K.pneumoniae for were 12h static effect 16.1±10; -1 log drop 22.8±12.5; -2 log drop 32.4±12.4; chemotherapy for more than forty years yet the pre-clinical data on their plazomicin administered 12hrly pharmacokinetics-pharmacodynamics (PK-PD) is scarce. -3 log drop 59.3±11.9; 24h static effect 49.5±12.7; -1 log drop 55.7±14.8; -2 Endpoint strain AUC/MIC Published data points to AUC/MIC or Cmax/MIC as the dominant log drop 64.1±19.5; -3 log drop 73.3±25.3; 48hr static effect 78.6±35.9; -1 AKPN 1169 AKPN 1170 AKPN 1171 KP41965 KP41966 Mean ± STD pharmacodynamic index (PDI) with an AUC/MIC of 50-70 being associated log drop 81.0±36.6; -2 log drop 81.8±37.8; -3 log drop 84.2±38.9. 12h with 24h static effect for aerobic Gram-negative rods ( Enterobacteriaceae The AUC/MIC targets for plazomicin and amikacin against E.coli when 24h Static 15.2 66.1 40.7 37.6 17.2 35.3±20.7 -1 log drop 28.8 77.6 49.6 59.3 23.7 47.8±22.2 and Pseudomonas aeruginosa ).
    [Show full text]
  • Recarbrio, INN-Imipenem / Cilastatin / Relebactam
    EMA/572792/2020 EMEA/H/C/004808 Recarbrio (imipenem / cilastatin / relebactam) An overview of Recarbrio and why it is authorised in the EU What is Recarbrio and what is it used for? Recarbrio is an antibiotic for treating adults with the following infections: • lung infections caught in hospital (hospital-acquired pneumonia), including ventilator-associated pneumonia (pneumonia caught while on a ventilator, which is a machine that helps a patient to breathe); • infection that has spread into the blood (bacteraemia) as a likely complication of hospital-acquired pneumonia or ventilator-associated pneumonia; • infections caused by bacteria classed as aerobic Gram-negative bacteria when other treatments might not work. Official guidance on the appropriate use of antibiotics should be considered when using the medicine. Recarbrio contains the active substances imipenem, cilastatin and relebactam. How is Recarbrio used? Recarbrio can only be obtained with a prescription and it should be used only after consulting a doctor with experience of managing infectious diseases. Recarbrio is given by infusion (drip) into a vein over 30 minutes. It is given every 6 hours for 5 to 14 days, depending on the nature of the infection. For more information about using Recarbrio, see the package leaflet or contact your doctor or pharmacist. How does Recarbrio work? One of the active substances in Recarbrio, imipenem, kills bacteria and the other two, cilastatin and relebactam, increase imipenem’s effectiveness in different ways. Imipenem interferes with bacterial proteins that are important for building the bacterial cell wall. This results in defective cell walls that collapse and cause the bacteria to die.
    [Show full text]
  • New Β-Lactamase Inhibitor Combinations: Options for Treatment; Challenges for Testing
    MEDICAL/SCIENTIFIC AffAIRS BULLETIN New β-lactamase Inhibitor Combinations: Options for Treatment; Challenges for Testing Background The β-lactam class of antimicrobial agents has played a crucial role in the treatment of infectious diseases since the discovery of penicillin, but β–lactamases (enzymes produced by the bacteria that can hydrolyze the β-lactam core of the antibiotic) have provided an ever expanding threat to their successful use. Over a thousand β-lactamases have been described. They can be divided into classes based on their molecular structure (Classes A, B, C and D) or their function (e.g., penicillinase, oxacillinase, extended-spectrum activity, or carbapenemase activity).1 While the first approach to addressing the problem ofβ -lactamases was to develop β-lactamase stable β-lactam antibiotics, such as extended-spectrum cephalosporins, another strategy that has emerged is to combine existing β-lactam antibiotics with β-lactamase inhibitors. Key β-lactam/β-lactamase inhibitor combinations that have been used widely for over a decade include amoxicillin/clavulanic acid, ampicillin/sulbactam, and pipercillin/tazobactam. The continued use of β-lactams has been threatened by the emergence and spread of extended-spectrum β-lactamases (ESBLs) and more recently by carbapenemases. The global spread of carbapenemase-producing organisms (CPOs) including Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii, limits the use of all β-lactam agents, including extended-spectrum cephalosporins (e.g., cefotaxime, ceftriaxone, and ceftazidime) and the carbapenems (doripenem, ertapenem, imipenem, and meropenem). This has led to international concern and calls to action, including encouraging the development of new antimicrobial agents, enhancing infection prevention, and strengthening surveillance systems.
    [Show full text]
  • AMEG Categorisation of Antibiotics
    12 December 2019 EMA/CVMP/CHMP/682198/2017 Committee for Medicinal Products for Veterinary use (CVMP) Committee for Medicinal Products for Human Use (CHMP) Categorisation of antibiotics in the European Union Answer to the request from the European Commission for updating the scientific advice on the impact on public health and animal health of the use of antibiotics in animals Agreed by the Antimicrobial Advice ad hoc Expert Group (AMEG) 29 October 2018 Adopted by the CVMP for release for consultation 24 January 2019 Adopted by the CHMP for release for consultation 31 January 2019 Start of public consultation 5 February 2019 End of consultation (deadline for comments) 30 April 2019 Agreed by the Antimicrobial Advice ad hoc Expert Group (AMEG) 19 November 2019 Adopted by the CVMP 5 December 2019 Adopted by the CHMP 12 December 2019 Official address Domenico Scarlattilaan 6 ● 1083 HS Amsterdam ● The Netherlands Address for visits and deliveries Refer to www.ema.europa.eu/how-to-find-us Send us a question Go to www.ema.europa.eu/contact Telephone +31 (0)88 781 6000 An agency of the European Union © European Medicines Agency, 2020. Reproduction is authorised provided the source is acknowledged. Categorisation of antibiotics in the European Union Table of Contents 1. Summary assessment and recommendations .......................................... 3 2. Introduction ............................................................................................ 7 2.1. Background ........................................................................................................
    [Show full text]
  • ZEMDRI (Plazomicin) Injection, for Intravenous Use (Ml/Min) ZEMDRI B Dosing Interval Initial U.S
    HIGHLIGHTS OF PRESCRIBING INFORMATION Recommended initial dosage regimen for patients with renal These highlights do not include all the information needed to use impairment is shown in the table below. (2.3) ZEMDRI safely and effectively. See full prescribing Recommended information for ZEMDRI. Estimated CLcr a Dosage for ZEMDRI (plazomicin) injection, for intravenous use (mL/min) ZEMDRI b Dosing Interval Initial U.S. Approval: 2018 Greater than or equal to 60 15 mg/kg Every 24 hours WARNING: NEPHROTOXICITY, OTOTOXICITY, to less than 90 NEUROMUSCULAR BLOCKADE and FETAL HARM Greater than or equal to 30 10 mg/kg Every 24 hours See full prescribing information for complete boxed warning. to less than 60 Nephrotoxicity has been reported with ZEMDRI. The risk Greater than or equal to 15 10 mg/kg Every 48 hours of nephrotoxicity is greater in patients with impaired renal to less than 30 a CLcr estimated by the Cockcroft-Gault formula. (2.3) function, the elderly, and in those receiving concomitant b nephrotoxic medications. (5.1) Calculate dosage using Total Body Weight (TBW). For patients Ototoxicity, manifested as hearing loss, tinnitus, and/or with TBW greater than IBW by 25% or more, use adjusted body weight. (2.3) vertigo, has been reported with ZEMDRI. Symptoms of aminoglycoside associated ototoxicity may be irreversible See Full Prescribing Information for subsequent dosage and may not become evident until after completion of adjustment based on changes in renal function or Therapeutic therapy. (5.2) Drug Monitoring (TDM). (2.3, 2.4). Aminoglycosides have been associated with neuromuscular See Full Prescribing Information for instructions on preparation blockade.
    [Show full text]
  • Prospects for Circumventing Aminoglycoside Kinase Mediated Antibiotic Resistance
    REVIEW ARTICLE published: 25 June 2013 CELLULAR AND INFECTION MICROBIOLOGY doi: 10.3389/fcimb.2013.00022 Prospects for circumventing aminoglycoside kinase mediated antibiotic resistance Kun Shi 1, Shane J. Caldwell 1, Desiree H. Fong 1* and Albert M. Berghuis 1,2* 1 Groupe de Recherche Axé sur la Structure des Protéines, Department of Biochemistry, McGill University, Montreal, QC, Canada 2 Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada Edited by: Aminoglycosides are a class of antibiotics with a broad spectrum of antimicrobial activity. Marcelo Tolmasky, California state Unfortunately, resistance in clinical isolates is pervasive, rendering many aminoglycosides University Fullerton, USA ineffective. The most widely disseminated means of resistance to this class of antibiotics Reviewed by: is inactivation of the drug by aminoglycoside-modifying enzymes (AMEs). There are two Sylvie Garneau-Tsodikova, University of Michigan, USA principal strategies to overcoming the effects of AMEs. The first approach involves the Maria S. Ramirez, IMPaM design of novel aminoglycosides that can evade modification. Although this strategy has (UBA-CONICET), Argentina yielded a number of superior aminoglycoside variants, their efficacy cannot be sustained in *Correspondence: the long term. The second approach entails the development of molecules that interfere Desiree H. Fong, Groupe de with the mechanism of AMEs such that the activity of aminoglycosides is preserved. Recherche Axé sur la Structure des Protéines, Department of Although such a molecule has yet to enter clinical development, the search for AME Biochemistry, McGill University, inhibitors has been greatly facilitated by the wealth of structural information amassed 3649 Promenade Sir William Osler, in recent years.
    [Show full text]
  • E200069A Sihuan Pharm 1..2
    Hong Kong Exchanges and Clearing Limited and The Stock Exchange of Hong Kong Limited take no responsibility for the contents of this announcement, make no representation as to its accuracy or completeness and expressly disclaim any liability whatsoever for any loss howsoever arising from or in reliance upon the whole or any part of the contents of this announcement. Sihuan Pharmaceutical Holdings Group Ltd. 四環醫藥控股集團有限公司 (incorporated in Bermuda with limited liability) (Stock code: 0460) VOLUNTARY ANNOUNCEMENT ACQUISITION OF ALL INTERESTS AND INTELLECTUAL PROPERTY RIGHTS OF PLAZOMICIN, A NEW GENERATION OF AMINOGLYCOSIDE ANTIBIOTICS IN THE GREATER CHINA REGION The board of directors (the ‘‘Board’’) of Sihuan Pharmaceutical Holdings Group Ltd. (the ‘‘Company’’ or ‘‘Sihuan Pharmaceutical’’, together with its subsidiaries, the ‘‘Group’’) is pleased to announce that Xuanzhu (HK) Biopharmaceutical Limited (‘‘Xuanzhu’’), a wholly-owned subsidiary of the Group, acquired all interests and intellectual property rights of plazomicin, a new generation of aminoglycoside antibiotics, in the Greater China Region (including the People’s Republic of China (the ‘‘PRC’’), Hong Kong Special Administrative Region, Macau Special Administrative Region and Taiwan) from Achaogen, Inc. (‘‘Achaogen’’), a company incorporated in Delaware, the United States of America. Plazomicin is a new generation of semisynthetic aminoglycoside antibiotics developed by Achaogen. It produces antibacterial effect mainly through binding to bacterial 30S ribosomal subunits and is used to treat severe infections caused by multi-drug resistance (‘‘MDR’’) gram-negative bacteria and enterobacteriaceae, including the carbapenem-resistant enterobacteriaceae. In the PRC, MDR bacterial infections are mainly acquired in hospital and most of the infected individuals are immunocompromised due to various reasons.
    [Show full text]
  • Title. 1 in Vitro Activity of the New Β-Lactamase Inhibitors Relebactam
    bioRxiv preprint doi: https://doi.org/10.1101/499830; this version posted December 19, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Title. 2 In vitro activity of the new β-lactamase inhibitors relebactam and vaborbactam in combination 3 with β-lactams against Mycobacterium abscessus complex clinical isolates 4 5 Authors and affiliations. Amit Kaushik,a Nicole C. Ammerman,a Jin Lee,a Olumide Martins,a 6 Barry N. Kreiswirth,b Gyanu Lamichhane,a Nicole M. Parrish,c Eric L. Nuermbergera 7 8 aCenter for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, 9 Maryland, USA 10 bPublic Health Research Institute Tuberculosis Center, New Jersey Medical School - Rutgers, 11 The State University of New Jersey, Newark, New Jersey, USA 12 cDepartment of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 13 USA 14 15 Key words. β-lactamase inhibitors, β-lactams, relebactam, vaborbactam, carbapenems, 16 cephalosporins, Mycobacterium abscessus 17 18 Running title. Relebactam/vaborbactam with β-lactams vs. M. abscessus Page 1 bioRxiv preprint doi: https://doi.org/10.1101/499830; this version posted December 19, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 19 Abstract 20 Pulmonary disease due to infection with Mycobacterium abscessus complex (MABC) is 21 notoriously difficult to treat, in large part due to MABC’s intrinsic resistance to most antibiotics, 22 including β-lactams.
    [Show full text]
  • Plazomicin for Complicated Urinary Tract Infection
    October Horizon Scanning Research & 2016 Intelligence Centre Plazomicin for complicated urinary tract infection NIHR HSRIC ID: 9787 Lay summary Serious infections caused by Gram-negative bacteria are becoming increasingly resistant to commonly prescribed antibiotics and are a serious global concern. If licensed, plazomicin will offer a treatment option for those patients who have a complicated urinary tract infection or acute pyelonephritis caused by multi-drug resistant Gram-negative bacteria, a group who currently have few effective and well tolerated therapies available. This briefing is based on information available at the time of research and a limited literature search. It is not intended to be a definitive statement on the safety, efficacy or effectiveness of the health technology covered and should not be used for commercial purposes or commissioning without additional information. This briefing presents independent research funded by the National Institute for Health Research (NIHR). The views expressed are those of the author and not necessarily those of the NHS, the NIHR or the Department of Health. Horizon Scanning Research & Intelligence Centre University of Birmingham [email protected] www.hsric.nihr.ac.uk @OfficialNHSC TARGET GROUP • Complicated urinary tract infection (cUTI), including acute pyelonephritis (AP); infection caused by resistant Gram-negative bacterial pathogens, including 3rd generation cephalosporin and carbapenem-resistant enterobacteriaceae (CRE) – first line, followed by appropriate, optional oral step down therapy. TECHNOLOGY DESCRIPTION Plazomicin (ACHN-490) is a next-generation broad-spectrum aminoglycoside antibiotic. Aminoglycosides kill bacteria by inhibiting protein synthesis through binding to the bacterial 16S rRNA, and by disrupting the integrity of bacterial cell membranes.
    [Show full text]
  • Nationwide Epidemiology of Carbapenem Resistant Klebsiella
    Galani et al. BMC Infectious Diseases (2019) 19:167 https://doi.org/10.1186/s12879-019-3801-1 RESEARCH ARTICLE Open Access Nationwide epidemiology of carbapenem resistant Klebsiella pneumoniae isolates from Greek hospitals, with regards to plazomicin and aminoglycoside resistance Irene Galani1,3*† , Konstantina Nafplioti1†, Panagiota Adamou1, Ilias Karaiskos2, Helen Giamarellou2, Maria Souli1 and Study Collaborators Abstract Background: To evaluate the in vitro activities of plazomicin and comparator aminoglycosides and elucidate the underlying aminoglycoside resistance mechanisms among carbapenemase-producing K. pneumoniae isolates collected during a nationwide surveillance study in Greek hospitals. Methods: Three hundred single-patient carbapenemase-producing K. pneumoniae isolates were studied, including 200 KPC-, 50 NDM-, 21 VIM-, 14 KPC & VIM-, 12 OXA-48-, two NDM & OXA- and one KPC & OXA-producing isolates. Susceptibility testing was performed by broth microdilution, and minimum inhibitory concentrations (MICs) interpreted per EUCAST breakpoints. Carbapenemase-, aminoglycoside modifying enzyme- and 16S rRNA methylase- encoding genes were detected by PCR. Results: Of 300 isolates tested, 5.7% were pandrug resistant and 29.3% extensively drug resistant. Plazomicin inhibited 87.0% of the isolates at ≤2 mg/L, with MIC50/MIC90 of 0.5/4 mg/L. Apramycin (a veterinary aminoglycoside) inhibited 86.7% of the isolates at ≤8 mg/L and was the second most active drug after plazomicin, followed by gentamicin (S, 43%; MIC50/MIC90, 4/> 256) and amikacin (S, 18.0%; MIC50/MIC90, 32/128). Twenty-three (7.7%) isolates (16 KPC-, 6 VIM- and one KPC & OXA-48-producers) exhibited MICs ≥64 mg/L for plazomicin, and harbored rmtB (n = 22) or armA (n = 1).
    [Show full text]