Advances in the Management of Myeloma

Total Page:16

File Type:pdf, Size:1020Kb

Advances in the Management of Myeloma Advances in the Management of Myeloma Parameswaran Hari, MD Armand J. Quick/William F. Stapp Professor of Hematology Director, Adult Blood and Marrow Transplant Program Froedtert Hospital Medical College of Wisconsin Milwaukee, WI Induction/Transplant/Maintenance- what does it get us? UPDATED OS DATA from CALGB 100104 & IFM 2005-02 REF:https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm542791.htm Quest for a Better Initial Therapy for MM KRD-Dara vs KRD Outcome (%) After 4 Cycles After 8 Cycles KRD-DARA KRD KRD-DARA KRD N = 21 Non-SCT N = 15 Non-SCT N = 49 N = 44 CR/sCR 5% 18% 27% 34% ≥VGPR 71% 69% 87% 89% Judging with limited data is fraught with error Evolution CyBorD 41%> VGPR 53% > VGPR 1 year PFS -100% Kumar RVD 32%> VGPR 51%>VGPR 1 year PFS - 83% CVRD 33%> VGPR 58%>VGPR 1year PFS - 86% Jakubowiak A et al. ASCO 2017, Abstract 8000. Jakubowiak, A et al. Blood 2012;120:1801-9. Kumar et al. Blood. 2012 May 10;119(19):4375 MM Risk Categories Standard Risk (80%) High Risk (20%) Risk Factors (Expected OS: 6-7 Yrs) (Expected OS: 2-3 Yrs) del(17p), t(4;14)* FISH t(11;14), t(6;14) t(14;16), +1q21 Hypodiploidy Cytogenetics Hyperdiploidy del(13q) β2-microglobulin* Low (< 3.5 mg/L) High (≥ 5.5 mg/L) PCLI < 3% High (≥ 3%) Gene expression profile Good risk High risk • Other high risk features: − Extramedullary disease − Plasma cell leukemia − Plasmablastic morphology *Patients with t(4;14), β2-microglobulin < 4 mg/L, and Hb ≥ 10 g/dL may have intermediate-risk disease. Dispenzieri A, et al. Mayo Clin Proc. 2007;82:323-341. Kumar SK, et al. Mayo Clin Proc. 2009;84:1095-1110. Mikhael JR, et al. Mayo Clin Proc. 2013;88:360-376. NCCN. Clinical practice guidelines in oncology: multiple myeloma. v.1.2015. Chng WJ, et al. Leukemia. 2014;28:269-277. Phase III Stamina Trial— BMT CTN0702 Lenalidomide Maintenance** Any induction therapy MEL 200mg/m2 Lenalidomide VRD x 4* Randomize Maintenance** ASCT 2 MEL 200mg/m Lenalidomide ASCT Maintenance** * Bortezomib 1.3mg /m2 days 1, 4, 8,11 Lenalidomide 15mg days 1-15 Dexamethasone 40mg days 1, 8, 15 **Lenalidomide 15 mg daily x 3years • Estimated study completion date: 2020 • Estimated primary initial completion date: 5/2016 ClinicalTrials.gov Identifier: NCT01109004 BMT CTN0702 STaMINA Trial Results Post induction + R Maint only RVD→R Double ASCT-1 followed by: n=257 n=254 ASCT→R n=247 Median PFS, mos 52.2 56.7 56.5 Median OS, mos 83.4 85.7 82.0 High-risk patients, n 59 65 57 Median PFS, 40.2 48.3 42.2 mos Median OS, 79.5 77.5 79.3 mos No significant difference between the study arms ClinicalTrials.gov. NCT01109004. VRd Maintenance After ASCT in High Risk Disease • 45 patients received VRd maintenance after ASCT for 2 years − Bortezomib 1.3mg/m2 weekly − Lenalidomide 10mg d1-21 − Dexamethasone 40mg weekly. 100 High-risk Features n (%) 90 sCR% Del 17p 19 (42) 80 sCR+CR % Del 1p 9 (20) 70 ≥ VGPR % T (4;14) 2 (5) 60 ORR % T (14;16) 5 (11) 50 SD % PD % PCL 11 (24) 40 30 Others (aggressive presentation) 7 (16) 20 > 1 Cytogenetic abnormalities 34 (75) 10 0 Post-induction Post-ASCT day 60 Best PFS: 32 months Response Response Response 3-year OS: 93% from: Nooka, AK, et al. Leukemia.2014 28,690-693 Beyond First Trial– Ongoing Studies For Newly Diagnosed MM Patients Ineligible For Transplant Sponsor/Study Study Intervention Status Director Eloquent-1 Bristol-Myers Ongoing, but not Len/dex +/- Elotuzumab NCT01335399 Squibb/AbbVie recruiting Janssen Len/dex +/- MAIA Trial Research & Currently Daratumumab NCT02252172 Development, recruiting LLC Len/dex +/- KEYNOTE-185 Merck Sharp & Currently Pembrolizumab NCT02579863 Dohme Corp. recruiting Emerging Proteasome-inhibitor─based Regimens For Transplant Ineligible Patients Study n ≥ VGPR ORR Ixazomib, Lenalidomide, dex x12 65 58% 95% Ixazomib maintenance[1] Ixazomib, Cyclophosphamide 300/400, dex x13 80/73% 70 27/23% Ixazomib maintenance[2] Carfilzomib*, Cyclophosphamide, dex x9 58 71% 95% Carfilzomib maintenance[3] Weekly Carfilzomib**, Cyclophosphamide, dex x9 47 87% 87% Carfilzomib maintenance[4] *twice a week dosing 20mg/m2 on day 1, 2 and 36mg/m2 on day 8 and afterwards **weekly dosing 70 mg/m2 on days 1, 8, 15 1 Kumar SK, et al. Lancet Oncol. 2014;15:1503-1512, 2. Dimopoulos MA, et al. ASH 2015. Abstract 26. 3. Bringhen S, et al. Blood. 2014;124:63-69, 4. Palumbo A , et al. ASH 2014. Abstract 175. MODERN TRIPLETS FOR RELAPSE Carfilzomib – Len – Dex Ixazomib – Len – Dex Elotuzumab – Len –Dex Daratumumab – Len -Dex Bortezomib – Panabinostat – Dex Bortezomib - Daratumumab- Dex ASPIRE—Len/Dex ± Carfilzomib in R/R MM: PFS 1.0 Median PFS: Carfilzomib (KRd): 26.3 months 0.8 Control Group (Rd): 17.6 months 0.6 0.4 up 0.2 0.0 HR 0.69 (95%CI, 0.57-0.83) P < .0001 Proportion Proportion Surviving w/o Progression 0 6 12 18 24 30 36 42 48 Months Since Randomization Risk Group by KRd (n = 396) Rd (n = 396) FISH HR P Value n Median PFS, Mos n Median PFS, Mos High 48 23.1 52 13.9 0.70 .083 Standard 147 29.6 170 19.5 0.66 .004 13 Stewart , et al. N Engl J Med. 2015;372:142-152. ELOQUENT-2—Len/Dex ± Elotuzumab in R/R MM: PFS 14 Figure from Lonial, S. et al. N Engl J Med. 2015; 373:621-663. TOURMALINE-MM1— Len/Dex ± Ixazomib: PFS Median PFS: 1.0 IRd: 20.6 months Placebo-Rd: 14.7 months 0.8 free survival free - 0.6 0.4 0.2 Log-rank test P=.012 Hazard ratio (95% CI): 0.742 (0.587, 0.939) Number of events: IRd 129; placebo-Rd 157 Probability progression of Probability 0.0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 Number of patients at risk: Time from randomization (months) IRd 360 345 332 315 298 283 270 248 233 224 206 182 145 119 111 95 72 58 44 34 26 14 9 1 0 Placebo-Rd 362 340 325 308 288 274 254 237 218 208 188 157 130 101 85 71 58 46 31 22 15 5 3 0 0 • Median number of cycles IRD 13 (1-26) vs 12 Rd (1-25) • 55% (IRd) and 52% (IR) of patients remain on treatment Moreau, P. et al. ASH 2015 Abstract 727 15 Summary of PFS 30 26.3 25 20.6 20 19.4 17.6 14.9 14.7 15 Rd 10 KRd IRd EloRd 5 0 ASPIRE TOURMALINE ELOQUENT-2 Trial Design PFS Invest. arm PFS Rd arm HR ASPIRE Rd vs KRd 26.3mo 17.6mo 0.69 TOURMALINE Rd vs IRd 19.4mo 14.9mo 0.70 ELOUQUENT-2 Rd vs EloRd 20.6mo 14.7mo 0.74 Moreau, P. et al. ASH 2015 Abstract 727 Lonial, S. et al. N Engl J Med. 2015; 373:621-663. 16 Stewart, et al. N Engl J Med. 2015;372:142-150. MAb-Based Targeting of Myeloma Antibody-dependent Complement-dependent Apoptosis/growth cellular cytotoxicity (ADCC) cytotoxicity (CDC) arrest via targeting C1q signaling pathways Effector C1q cells: CDC MM MM FcR Daratumumab (CD38) Daratumumab (CD38) SAR650984 (CD38) SAR650984 (CD38) ADCC MM Elotuzumab (SLAMF7) Daratumumab (CD38) SAR650984 (CD38) Tai YT, et al. Bone Marrow Res. 2011;2011:924058. CD38 Antibodies – Immune Effects CASTOR: Study Design Multicenter, randomized, open-label, active-controlled phase 3 study Key eligibility criteria Primary Endpoint R DVd (n = 251) Daratumumab (16 mg/kg IV) • RRMM • PFS A Every week - cycle 1-3 • ≥1 prior line of Every 3 weeks - cycle 4-8 Secondary Endpoints N Every 4 weeks - cycles 9+ therapy D • TTP Vel: 1.3 mg/m2 SC, days 1,4,8,11 - cycle 1-8 • OS • Prior bortezomib O dex: 20 mg PO-IV, days 1,2,4,5,8,9,11,12 - cycle 1-8 exposure, but not M 1:1 • ORR, VGPR, CR refractory I • MRD Vd (n = 247) Z • Time to response E Vel: 1.3 mg/m2 SC, days 1,4,8,11 - cycle 1-8 • Duration of response dex: 20 mg PO-IV, days 1,2,4,5,8,9,11,12 - cycle 1-8 • Cycles 1-8: repeat every 21 days • Cycles 9+: repeat every 28 days Daratumumab IV administered in 1000 mL to 500 mL; gradual escalation from 50 mL to 200 mL/min permitted RRMM, relapsed or refractory multiple myeloma; DVd, daratumumab/bortezomib/dexamethasone; IV, intravenous; Vel, bortezomib; SC, subcutaneous; dex, dexamethasone; PO, oral; Vd, bortezomib/dexamethasone; PFS, progression-free survival; TTP, time to progression; ORR, overall response rate; VGPR, very good partial response; CR, complete response; MRD, minimal residual disease. 19 Progression-free Survival 1.0 1-year PFS* Median : not reached 0.8 60.7% 0.6 DVd 0.4 26.9% 0.2 Median : 7.2 months Vd HR: 0.39 (95% CI, 0.28-0.53); P<0.0001 Proportion Proportion surviving without progression 0 0 3 6 9 12 15 No. at risk Months Vd 247 182 106 25 5 0 DVd 251 215 146 56 11 0 61% reduction in the risk of disease progression or death for DVd vs Vd *KM estimate; HR, hazard ratio. POLLUX – LENALIDOMIDE + DARA POLLUX STUDY 4 Major Triplets for Relapsed MM Do we know which triplet is better? KRD IRD ERD DRD DVD DPD ROLE OF RETRANSPLANTATION Second Transplant at Relapse Myeloma X: Salvage Transplant at Relapse Randomized 1:1 Melphalan 200mg/m2 IV + ASCT R/R MM; PAD induction (n = 89) >18 mos after prior ASCT 2-4 cycles (N = 293) Cyclophosphamide 400mg/m2 PO/wk x12 cycles (n = 85) Cook G et al; Lancet Haematol.
Recommended publications
  • Predictive QSAR Tools to Aid in Early Process Development of Monoclonal Antibodies
    Predictive QSAR tools to aid in early process development of monoclonal antibodies John Micael Andreas Karlberg Published work submitted to Newcastle University for the degree of Doctor of Philosophy in the School of Engineering November 2019 Abstract Monoclonal antibodies (mAbs) have become one of the fastest growing markets for diagnostic and therapeutic treatments over the last 30 years with a global sales revenue around $89 billion reported in 2017. A popular framework widely used in pharmaceutical industries for designing manufacturing processes for mAbs is Quality by Design (QbD) due to providing a structured and systematic approach in investigation and screening process parameters that might influence the product quality. However, due to the large number of product quality attributes (CQAs) and process parameters that exist in an mAb process platform, extensive investigation is needed to characterise their impact on the product quality which makes the process development costly and time consuming. There is thus an urgent need for methods and tools that can be used for early risk-based selection of critical product properties and process factors to reduce the number of potential factors that have to be investigated, thereby aiding in speeding up the process development and reduce costs. In this study, a framework for predictive model development based on Quantitative Structure- Activity Relationship (QSAR) modelling was developed to link structural features and properties of mAbs to Hydrophobic Interaction Chromatography (HIC) retention times and expressed mAb yield from HEK cells. Model development was based on a structured approach for incremental model refinement and evaluation that aided in increasing model performance until becoming acceptable in accordance to the OECD guidelines for QSAR models.
    [Show full text]
  • Clinical Use of Proteasome Inhibitors in the Treatment of Multiple Myeloma
    Pharmaceuticals 2015, 8, 1-20; doi:10.3390/ph8010001 OPEN ACCESS pharmaceuticals ISSN 1424-8247 www.mdpi.com/journal/pharmaceuticals Review Clinical Use of Proteasome Inhibitors in the Treatment of Multiple Myeloma Noah M. Merin and Kevin R. Kelly * Internal Medicine, Jane Anne Nohl Division of Hematology, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-323-865-3823; Fax: +1-323-865-0060. Academic Editor: Jennifer Carew Received: 29 July 2014 / Accepted: 4 December 2014 / Published: 24 December 2014 Abstract: Multiple myeloma (MM) is an incurable hematological malignancy characterized by the clonal proliferation of neoplastic plasma cells. The use of proteasome inhibitors in the treatment of MM has led to significant improvements in outcomes. This article reviews data on the use of the two approved proteasome inhibitors (bortezomib and carlfilzomib), as well as newer agents under development. Emphasis is placed on the clinical use of proteasome inhibitors, including management of side effects and combination with other agents. Keywords: multiple myeloma; proteasome inhibitors; toxicity 1. Introduction Multiple myeloma (MM) is a malignant neoplasm characterized by clonal proliferation of plasma cells in the bone marrow that produce monoclonal protein (M-protein) that may be detected in the serum or urine. It is the second most common hematologic cancer in the United States (after Non-Hodgkin Lymphoma); an estimated 24,050 new cases and 11,090 deaths will occur due to MM in the USA in 2014 [1].
    [Show full text]
  • A Phase 1 Dose-Escalation Study of Filanesib Plus
    A Phase 1 Dose-Escalation Study of Filanesib Plus Bortezomib and Dexamethasone in Patients With Recurrent/ Refractory Multiple Myeloma Ajai Chari, Icahn School of Medicine at Mount Sinai Myo Htut, City Hope National Medical Center Jeffrey A. Zonder, Wayne State University Joseph W. Fay, Baylor Research Institute Andrzej Jakubowiak, University of Chicago Joan B. Levy, Multiple Myeloma Research Consortium Kenneth Lau, Icahn School of Medicine at Mount Sinai Steven M. Burt, Wayne State University Brian J. Tunquist, Array BioPharma Inc Brandi W. Hilder, Array BioPharma Inc Only first 10 authors above; see publication for full author list. Journal Title: Cancer Volume: Volume 122, Number 21 Publisher: Wiley | 2016-11-01, Pages 3327-3335 Type of Work: Article | Post-print: After Peer Review Publisher DOI: 10.1002/cncr.30174 Permanent URL: https://pid.emory.edu/ark:/25593/vj1mj Final published version: http://dx.doi.org/10.1002/cncr.30174 Copyright information: © 2016 American Cancer Society Accessed October 1, 2021 7:46 AM EDT HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Cancer. Manuscript Author Author manuscript; Manuscript Author available in PMC 2019 November 15. Published in final edited form as: Cancer. 2016 November 15; 122(21): 3327–3335. doi:10.1002/cncr.30174. A Phase 1 Dose-Escalation Study of Filanesib Plus Bortezomib and Dexamethasone in Patients With Recurrent/Refractory Multiple Myeloma Ajai Chari, MD1, Myo Htut, MD2, Jeffrey A. Zonder, MD3, Joseph W. Fay, MD4, Andrzej J. Jakubowiak, MD5, Joan B. Levy, PhD6, Kenneth Lau, BA1, Steven M. Burt, MSN, NP-C3, Brian J. Tunquist, PhD7, Brandi W. Hilder, PhD7, Selena A.
    [Show full text]
  • ADC (Antibody-Drug Conjugates
    Antibody-drug Conjugate All-sided compounds and intermediates to support novel biotherapeutics research and technology Antibody- drug Conjugate ADC Products Based on advanced technology and years of experience, Creative Biolabs offers a variety of customized ADC, linker-Toxin ANTIBODY-DRUG products, ADCs cytotoxin, ADCs linkers, Anti-drug Abs and Anti-Ab ADCs for your CONJUGATE ADC projects and promote your progress in ADC development. Comprehensive one-stop-shop for all aspects in ADC research and development Creative Biolabs Established in 2004, Creative Biolabs is a world-renowned service provider for antibody-drug conjugate (ADC) service and products. After years of pursuit for perfection, Creative BioLabs has established leadership in targeted immunotherapy and ADC development. Outline 01.Customized ADCs 02.Linker-Toxin 03.ADCs Cytotoxin 04.ADCs Linker 05.Anti-drug Abs Creative Biolabs provides a 06.Anti-Ab ADCs full range of ADC products to help your 07.Contact us ADC development . Antibody- drug Conjugate CUSTOMIZED ADCS Creative Biolabs offers a variety of customarily prepared ADC products to serve the course of ADC development and evaluation. Taken advantage of our large inventory of therapeutic antibodies, our scientists help create the custom ADC products by conjugating therapeutic antibodies against cancer surface antigens with different drug-linker complexes. APPLICATION ADCs have created a new paradigm for novel anticancer drug development. With both the specificity of the large-molecule monoclonal antibody and the potency of the small molecule cytotoxic drug, ADCs have tremendous potential to be part of the future of cancer precision medicine as well as cancer combination therapies. Customized ADCs Cat.
    [Show full text]
  • Pomalidomide Enhanced Gemcitabine and Nab-Paclitaxel on Pancreatic Cancer Both in Vitro and in Vivo
    www.oncotarget.com Oncotarget, 2018, Vol. 9, (No. 21), pp: 15780-15791 Research Paper Pomalidomide enhanced gemcitabine and nab-paclitaxel on pancreatic cancer both in vitro and in vivo Nobuhiro Saito1,2, Yoshihiro Shirai1,2,*, Tadashi Uwagawa1,3, Takashi Horiuchi1,2, Hiroshi Sugano1,2, Koichiro Haruki1, Hiroaki Shiba1, Toya Ohashi2 and Katsuhiko Yanaga1 1Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan 2Division of Gene Therapy, Research Center for Medical Science, The Jikei University School of Medicine, Tokyo, Japan 3Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan *This author contributed equally to this work Correspondence to: Nobuhiro Saito, email: [email protected] Keywords: IMiDs; pancreatic cancer; gemcitabine/nab-paclitaxel; NF-κB; p53 Received: December 29, 2017 Accepted: February 25, 2018 Epub: March 02, 2018 Published: March 20, 2018 Copyright: Saito et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0 (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT Background: Chemotherapy with gemcitabine and nab-paclitaxel (gemcitabine/ nab-paclitaxel) is recommended for unresectable pancreatic cancer. However, the therapeutic efficacy is attenuated by the antitumor agent-induced activation of nuclear factor-κB (NF-κB). Thalidomide inhibits NF-κB activation, therefore, we hypothesized that pomalidomide, a third-generation IMiD, would also inhibit NF-κB activation and enhance the antitumor effects of gemcitabine/nab-paclitaxel. Methods: In vitro, we assessed NF-κB activity and apoptosis in response to pomalidomide alone, gemcitabine/nab-paclitaxel, or combination of pomalidomide and gemcitabine/nab-paclitaxel in human pancreatic cancer cell lines (PANC-1 and MIA PaCa-2).
    [Show full text]
  • Monoclonal Antibodies in Myeloma
    Monoclonal Antibodies in Myeloma Pia Sondergeld, PhD, Niels W. C. J. van de Donk, MD, PhD, Paul G. Richardson, MD, and Torben Plesner, MD Dr Sondergeld is a medical student at the Abstract: The development of monoclonal antibodies (mAbs) for University of Giessen in Giessen, Germany. the treatment of disease goes back to the vision of Paul Ehrlich in Dr van de Donk is a hematologist in the the late 19th century; however, the first successful treatment with department of hematology at the VU a mAb was not until 1982, in a lymphoma patient. In multiple University Medical Center in Amsterdam, The Netherlands. Dr Richardson is the R.J. myeloma, mAbs are a very recent and exciting addition to the Corman Professor of Medicine at Harvard therapeutic armamentarium. The incorporation of mAbs into Medical School, and clinical program current treatment strategies is hoped to enable more effective and leader and director of clinical research targeted treatment, resulting in improved outcomes for patients. at the Jerome Lipper Myeloma Center, A number of targets have been identified, including molecules division of hematologic malignancy, depart- on the surface of the myeloma cell and components of the bone ment of medical oncology, Dana-Farber Cancer Institute in Boston, MA. Dr Plesner marrow microenvironment. Our review focuses on a small number is a professor of hematology at the Univer- of promising mAbs directed against molecules on the surface of sity of Southern Denmark and a consultant myeloma cells, including CS1 (elotuzumab), CD38 (daratumumab, in the department of hematology at Vejle SAR650984, MOR03087), CD56 (lorvotuzumab mertansine), and Hospital in Vejle, Denmark.
    [Show full text]
  • Immunotherapies Shape the Treatment Landscape for Hematologic Malignancies Jane De Lartigue, Phd
    Feature Immunotherapies shape the treatment landscape for hematologic malignancies Jane de Lartigue, PhD he treatment landscape for hematologic of TIL therapy has been predominantly limited to malignancies is evolving faster than ever melanoma.1,3,4 before, with a range of available therapeutic Most recently, there has been a substantial buzz Toptions that is now almost as diverse as this group around the idea of genetically engineering T cells of tumors. Immunotherapy in particular is front and before they are reintroduced into the patient, to center in the battle to control these diseases. Here, increase their anti-tumor efficacy and minimize we describe the latest promising developments. damage to healthy tissue. This is achieved either by manipulating the antigen binding portion of the Exploiting T cells T-cell receptor to alter its specificity (TCR T cells) The treatment landscape for hematologic malig- or by generating artificial fusion receptors known as nancies is diverse, but one particular type of therapy chimeric antigen receptors (CAR T cells; Figure 1). has led the charge in improving patient outcomes. The former is limited by the need for the TCR to be Several features of hematologic malignancies may genetically matched to the patient’s immune type, make them particularly amenable to immunother- whereas the latter is more flexible in this regard and apy, including the fact that they are derived from has proved most successful. corrupt immune cells and come into constant con- CARs are formed by fusing part of the single- tact with other immune cells within the hemato- chain variable fragment of a monoclonal antibody poietic environment in which they reside.
    [Show full text]
  • Daratumumab (Darzalex)
    Clinical Advances in Immunotherapy in Myeloma 6/14/17 Webinar 1: Monoclonal Antibodies: A New Type of Myeloma Drug Clinical Advances in Immunotherapy in Myeloma Webinar 1, June 14, 2017 Monoclonal Antibodies Speakers Moderator: Mary DeRome Multiple Myeloma Research Foundation Norwalk, Connecticut Faculty: Tom Martin, MD University of California, San Francisco San Francisco, California Amy Marsala, MPH, MSN, NP University of California, San Francisco San Francisco, California Gary Rudman, CSP Myeloma Patient Advocate 1 Clinical Advances in Immunotherapy in Myeloma 6/14/17 Webinar 1: Monoclonal Antibodies: A New Type of Myeloma Drug Topics for Discussion • Current and future monoclonal antibodies (what they are, how they work) • Management of side effects in the era of monoclonal antibodies • Patient experience on phase 2 clinical trial of Empliciti (elotuzumab) Tom Martin, MD 2 Clinical Advances in Immunotherapy in Myeloma 6/14/17 Webinar 1: Monoclonal Antibodies: A New Type of Myeloma Drug Current Overall Survival for Newly Diagnosed Myeloma 1.0 89% vs 81% 0.9 Still losing 25% patients in first 3 years 75% vs 62% 0.8 0.7 2007–2011 0.6 0.5 0.4 0.3 Proportion Surviving 2001–2006 0.2 0.1 0.0 012345678910 Years From Diagnosis A good trend but we always need to improve!! Myeloma Treatment: It’s an Art 3 Clinical Advances in Immunotherapy in Myeloma 6/14/17 Webinar 1: Monoclonal Antibodies: A New Type of Myeloma Drug New Agents in Myeloma Therapy Oral Kinase HDAC Immuno- New IMiDs proteasomes inhibitors Novel MOA inhibitors therapies • Lenalidomide
    [Show full text]
  • Novel Targets in Multiple Myeloma
    · MULTIPLE MYELOMA · Novel Targets in Multiple Myeloma Douglas Tremblay, MD, and Ajai Chari, MD Abstract also being increasingly approved by the FDA as a part of combination Despite the availability of 6 classes of drugs for the therapy based on PFS extension of 3 to 6 months achieved in random- treatment of multiple myeloma (MM), the disease re- ized phase III studies. The goal of this review is to discuss the targets, mains incurable in most cases, with patients eventually mechanisms of action (MOA), safety, and efficacy of the novel agents relapsing on each of these agents. The benchmark for in MM that are currently have recently met or have the best chance of drug approval in heavily pretreated and multidrug-re- meeting these benchmarks. fractory patients is an overall response rate (ORR) of approximately 25% to 30% and progression-free Targets of Immunomodulatory Drugs survival (PFS) of approximately 4 months, but more Of note, there also have been developments in our understanding of recently in less heavily pretreated patients with relapsed the targets of immunomodulatory drugs (IMiDs). Until recently, it has disease after 1 to 3 lines of therapy, based primarily on been unclear how these oral medications achieve antimyeloma ben- an improvement in PFS of 3 to 6 months in random- efits without significant toxicity at the site of drug absorption in the ized phase III studies. This article reviews the targets, gut or other off-target organ toxicities. Recently, cereblon, a member mechanisms of action (MOA), safety, and efficacy of the of the E3 ubiquitin ligase family, was identified as the likely mediator recently approved agents in MM that have met one of of teratogenicity of IMiDs.
    [Show full text]
  • Salvage Therapies in Relapsed And/Or Refractory Myeloma: What Is Current and What Is the Future?
    Journal name: OncoTargets and Therapy Article Designation: Review Year: 2016 Volume: 9 OncoTargets and Therapy Dovepress Running head verso: Thumallapally et al Running head recto: Salvage therapies in relapsed and/or refractory myeloma open access to scientific and medical research DOI: http://dx.doi.org/10.2147/OTT.S110189 Open Access Full Text Article REVIEW Salvage therapies in relapsed and/or refractory myeloma: what is current and what is the future? Nishitha Thumallapally1 Abstract: The treatment landscape for multiple myeloma (MM) is evolving with our Hana Yu1 understanding of its pathophysiology. However, given the inevitable cohort heterogeneity Divya Asti1 in salvage therapy, response to treatment and overall prognoses tend to vary widely, making Adarsh Vennepureddy1 meaningful conclusions about treatment efficacy difficult to derive. Despite the hurdles in Terenig Terjanian2 current research, progress is underway toward more targeted therapeutic approaches. Several new drugs with novel mechanism of action and less toxic profile have been developed in the past 1Department of Internal Medicine, decade, with the potential for use as single agents or in synergy with other treatment modalities 2Division of Hematology and Oncology, Staten Island University in MM therapy. As our discovery of these emerging therapies progresses, so too does our need Hospital, New York, NY, USA to reshape our knowledge on knowing how to apply them. This review highlights some of the recent landmark changes in MM management with specific emphasis on salvage drugs available for relapsed and refractory MM and also discusses some of the upcoming cutting-edge therapies For personal use only. that are currently in various stages of clinical development.
    [Show full text]
  • Escalation Study of Filanesib Plus Bortezomib and Dexamethasone in Patients with Recurrent/Refractory Multiple Myeloma
    Original Article A Phase 1 Dose-Escalation Study of Filanesib Plus Bortezomib and Dexamethasone in Patients With Recurrent/Refractory Multiple Myeloma Ajai Chari, MD1; Myo Htut, MD2; Jeffrey A. Zonder, MD3; Joseph W. Fay, MD4; Andrzej J. Jakubowiak, MD5; Joan B. Levy, PhD6; Kenneth Lau, BA1; Steven M. Burt, MSN, NP-C3; Brian J. Tunquist, PhD7; Brandi W. Hilder, PhD7; Selena A. Rush, BS7; Duncan H. Walker, PhD7; Mieke Ptaszynski, MD7; and Jonathan L. Kaufman, MD8 BACKGROUND: Filanesib is a kinesin spindle protein inhibitor that has demonstrated encouraging activity in patients with recurrent/ refractory multiple myeloma. Preclinical synergy with bortezomib was the rationale for the current phase 1 study. METHODS: The cur- rent study was a multicenter study with an initial dose-escalation phase to determine the maximum tolerated dose of 2 schedules of filanesib plus bortezomib with and without dexamethasone, followed by a dose-expansion phase. RESULTS: With the addition of pro- phylactic filgastrim, the maximum planned dose was attained: 1.3 mg/m2/day of bortezomib plus 40 mg of dexamethasone on days 1, 8, and 15 of a 28-day cycle, with filanesib given intravenously either at a dose of 1.5 mg/m2/day (schedule 1: days 1, 2, 15, and 16) or 3 mg/m2/day (schedule 2: days 1 and 15). The most common adverse events (assessed for severity using version 4.0 of the National Cancer Institute Common Terminology Criteria for Adverse Events) were transient, noncumulative neutropenia and thrombocytopenia with grade 3/4 events reported in 44% (16% in cycle 1 with filgastrim) and 29% of patients, respectively.
    [Show full text]
  • Belantamab Mafodotin to Treat Multiple Myeloma: a Comprehensive Review of Disease, Drug Efficacy and Side Effects
    Review Belantamab Mafodotin to Treat Multiple Myeloma: A Comprehensive Review of Disease, Drug Efficacy and Side Effects Grace Lassiter 1,*, Cole Bergeron 2, Ryan Guedry 2, Julia Cucarola 2, Adam M. Kaye 3 , Elyse M. Cornett 4, Alan D. Kaye 4 , Giustino Varrassi 5, Omar Viswanath 4,6,7,8 and Ivan Urits 4,9 1 School of Medicine, Georgetown University, Washington, DC 20007, USA 2 School of Medicine, Louisiana State University Shreveport, Shreveport, LA 71103, USA; [email protected] (C.B.); [email protected] (R.G.); [email protected] (J.C.) 3 Department of Pharmacy Practice, Thomas J. Long School of Pharmacy and Health Sciences, University of the Pacific, Stockton, CA 95211, USA; akaye@pacific.edu 4 Department of Anesthesiology, Louisiana State University Shreveport, Shreveport, LA 71103, USA; [email protected] (E.M.C.); [email protected] (A.D.K.); [email protected] (O.V.); [email protected] (I.U.) 5 Paolo Procacci Foundation, Via Tacito 7, 00193 Roma, Italy; [email protected] 6 College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85724, USA 7 Department of Anesthesiology, School of Medicine, Creighton University, Omaha, NE 68124, USA 8 Valley Anesthesiology and Pain Consultants—Envision Physician Services, Phoenix, AZ 85004, USA 9 Southcoast Health, Southcoast Physicians Group Pain Medicine, Wareham, MA 02571, USA * Correspondence: [email protected] Received: 14 December 2020; Accepted: 18 January 2021; Published: 21 January 2021 Abstract: Multiple myeloma (MM) is a hematologic malignancy characterized by excessive clonal proliferation of plasma cells. The treatment of multiple myeloma presents a variety of unique challenges due to the complex molecular pathophysiology and incurable status of the disease at this time.
    [Show full text]