37 results on '"Lipe B"'
Search Results
2. Consensus guidelines and recommendations for infection prevention in multiple myeloma: a report from the International Myeloma Working Group
- Author
-
Raje, N.S. Anaissie, E. Kumar, S.K. Lonial, S. Martin, T. Gertz, M.A. Krishnan, A. Hari, P. Ludwig, H. O'Donnell, E. Yee, A. Kaufman, J.L. Cohen, A.D. Garderet, L. Wechalekar, A.F. Terpos, E. Khatry, N. Niesvizky, R. Yi, Q. Joshua, D.E. Saikia, T. Leung, N. Engelhardt, M. Mothy, M. Branagan, A. Chari, A. Reiman, A.J. Lipe, B. Richter, J. Rajkumar, S.V. Miguel, J.S. Anderson, K.C. Stadtmauer, E.A. Prabhala, R.H. McCarthy, P.L. Munshi, N.C.
- Abstract
Infection remains the leading cause of morbidity and mortality in patients with multiple myeloma because of the cumulative effect of disease, treatment, and host-related factors. Given that infectious risk is cumulative through the course of the disease, preventing infections is paramount. Optimal preventive strategies include vaccination against common pathogens, antimicrobial prophylaxis, infection control measures, and immunoglobulin replacement in a small subset of patients; however, there are no universally accepted guidelines for infection prevention. This Review provides a consensus statement from a panel of 36 experts with global representation, which was convened by The International Myeloma Society to review existing literature and current guidelines, address issues associated with the risk of infection and prevention of infectious complications in multiple myeloma in the context of emerging therapies, and offer recommendations for preventing these complications. © 2022 Elsevier Ltd
- Published
- 2022
3. PS1414 A PHASE 1B/2 STUDY OF SELINEXOR, CARFILZOMIB, AND DEXAMETHASONE (SKD) IN RELAPSED/REFRACTORY MULTIPLE MYELOMA (RRMM)
- Author
-
Gasparetto, C., primary, Lentzsch, S., additional, Schiller, G., additional, Callander, N., additional, Chen, C., additional, White, D., additional, Kotb, R., additional, Sutherland, H., additional, Sebag, M., additional, Tuchman, S., additional, Baljevic, M., additional, Bensinger, W., additional, LeBlanc, R., additional, Venner, C., additional, Bahlis, N., additional, Rodriguez-Lopez, K., additional, Sheehan, H., additional, Saint-Martin, J.-R., additional, Shah, J., additional, Shacham, S., additional, Kauffman, M., additional, and Lipe, B., additional
- Published
- 2019
- Full Text
- View/download PDF
4. PF587 SELINEXOR, POMALIDOMIDE, AND DEXAMETHASONE (SPD) IN PATIENTS WITH RELAPSED OR REFRACTORY MULTIPLE MYELOMA
- Author
-
Chen, C., primary, Gasparetto, C., additional, White, D., additional, Kotb, R., additional, Lipe, B., additional, Sutherland, H., additional, Sebag, M., additional, Tuchman, S., additional, Baljevic, M., additional, Bensinger, W., additional, LeBlanc, R., additional, Venner, C., additional, Lentzsch, S., additional, Schiller, G., additional, Callander, N., additional, Rodriguez-Lopez, K., additional, Sheehan, H., additional, Saint-Martin, J.-R., additional, Shah, J., additional, Shacham, S., additional, Kauffman, M., additional, and Bahlis, N., additional
- Published
- 2019
- Full Text
- View/download PDF
5. S1606 SAFETY AND EFFICACY OF COMBINATION OF SELINEXOR, DARATUMUMAB, AND DEXAMETHASONE (SDD) IN PATIENTS WITH MULTIPLE MYELOMA (MM) PREVIOUSLY EXPOSED TO PROTEASOME INHIBITORS AND IMMUNOMODULATORY DRUGS
- Author
-
Gasparetto, C., primary, Lentzsch, S., additional, Schiller, G., additional, Callander, N., additional, Tuchman, S., additional, Chen, C., additional, White, D., additional, Kotb, R., additional, Sutherland, H., additional, Sebag, M., additional, Baljevic, M., additional, Bensinger, W., additional, LeBlanc, R., additional, Venne, C., additional, Bahlis, N., additional, Rodriguez-Lopez, K., additional, Sheehan, H., additional, Saint-Martin, J.-R., additional, Shah, J., additional, Shacham, S., additional, Kauffman, M., additional, and Lipe, B., additional
- Published
- 2019
- Full Text
- View/download PDF
6. The role of maintenance therapy in multiple myeloma
- Author
-
Lipe, B, primary, Vukas, R, additional, and Mikhael, J, additional
- Published
- 2016
- Full Text
- View/download PDF
7. Phase I safety data of lenalidomide, bortezomib, dexamethasone, and elotuzumab as induction therapy for newly diagnosed symptomatic multiple myeloma: SWOG S1211
- Author
-
Usmani, S Z, primary, Sexton, R, additional, Ailawadhi, S, additional, Shah, J J, additional, Valent, J, additional, Rosenzweig, M, additional, Lipe, B, additional, Zonder, J A, additional, Fredette, S, additional, Durie, B, additional, Hoering, A, additional, Bartlett, B, additional, and Orlowski, R Z, additional
- Published
- 2015
- Full Text
- View/download PDF
8. Extracellular proteases in renal cell carcinoma.
- Author
-
Lipe, B., primary, Ernstoff, M. S., additional, Gui, J., additional, Seigne, J. D., additional, Schned, A., additional, and Petrella, B., additional
- Published
- 2011
- Full Text
- View/download PDF
9. Dose-ranging study of the combination of paclitaxel poliglumex and pemetrexed in advanced non-small cell lung cancer (NSCLC)
- Author
-
Slagle, B. M., primary, Rigas, J. R., additional, Dragnev, K. H., additional, Williams, I., additional, DiSalvo, W., additional, Engman, C., additional, Lipe, B., additional, and Simeone, S., additional
- Published
- 2009
- Full Text
- View/download PDF
10. Deficiencies of natural anticoagulants, protein C, protein s, and antithrombin.
- Author
-
Lipe B and Ornstein DL
- Published
- 2011
- Full Text
- View/download PDF
11. Clinical Management of Patients With Relapsed/Refractory Multiple Myeloma Treated With Talquetamab.
- Author
-
Chari A, Krishnan A, Rasche L, Ye JC, Garfall A, Popat R, Lipe B, Qin X, Campagna M, Masterson T, Tomlinson C, Hilder B, Tolbert J, Renaud T, Smit MD, Gray K, Kane C, Heuck C, and van de Donk NWCJ
- Subjects
- Humans, Disease Management, Male, Female, Multiple Myeloma drug therapy, Antibodies, Bispecific therapeutic use, Antibodies, Bispecific adverse effects, Antibodies, Bispecific pharmacology
- Abstract
Background: Talquetamab is a bispecific antibody targeting the multiple myeloma-associated antigen G protein-coupled receptor family C group 5 member D (GPRC5D). In the phase 1/2 MonumenTAL-1 trial (NCT03399799/NCT04634552), overall responses rates were > 71% in patients with triple-class exposed relapsed/refractory multiple myeloma (RRMM). Due to the distribution of the target antigen, a unique pattern of GPRC5D-associated adverse events (AEs) was observed, together with T-cell redirection-associated AEs. Management strategies for talquetamab-associated AEs are described., Discussion: GPRC5D-associated AEs included dermatologic (rash, nonrash, and nail toxicities) and oral AEs (dysgeusia, dysphagia, and dry mouth). The incidence of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were consistent with other T-cell redirection therapies. The incidence of high-grade infections was lower than observed with B-cell maturation antigen-targeting bispecific antibodies, with less frequent use of intravenous immunoglobulin required. GPRC5D-associated AEs were mostly low grade and led to few discontinuations. Skin toxicities were managed with emollients, topical corticosteroids, and oral corticosteroids (for high-grade, persistent, or AEs that progress). Nail toxicities were commonly managed with emollients. Based on investigator experience, dose modification may be effective for controlling oral events. Observation for potential weight changes is required. Infections were managed per standard of care. CRS and ICANS were effectively managed, consistent with other trials of T-cell redirection therapies., Conclusion: Although talquetamab had a distinct safety profile, AEs were considered clinically manageable and mostly low grade. With appropriate education and support, health care practitioners can ensure patients with RRMM maintain quality of life and treatment adherence. VIDEO ABSTRACT., (Copyright © 2024. Published by Elsevier Inc.)
- Published
- 2024
- Full Text
- View/download PDF
12. Personalized Treatment of Multiple Myeloma in Frail Patients.
- Author
-
Lipof JJ, Abdallah N, and Lipe B
- Subjects
- Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Frailty complications, Aged, Multiple Myeloma drug therapy, Multiple Myeloma therapy, Precision Medicine methods, Frail Elderly
- Abstract
Purpose of Review: As the treatment landscape for multiple myeloma (MM) continues to expand at a rapid pace, management of older adults and frail patients becomes increasingly challenging. As these patients have traditionally been underrepresented on clinical trials, there is limited guidance on the optimal approach to frail patients with newly diagnosed multiple myeloma (NDMM) or relapsed and refractory multiple myeloma (RRMM)., Recent Findings: Frailty is an independent predictor of tolerability and response to antineoplastic treatment. Stringent eligibility criteria have often excluded these patients, but recently some large trials have included frailty sub-analyses to help guide management. In general, triplet regimens are preferred to doublet regimens in this population and enrollment on a clinical trial should be prioritized when possible. In this review, we summarize the MM frailty scoring tools that have been developed to identify and assess this vulnerable population. We present the clinical trials over the past decade that have enrolled frail patients and/or have included subgroup analyses to help elucidate the response and tolerability of different regimens in this underrepresented group. We provide practical advice regarding assessment and management of frail patients NDMM and RRMM., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
13. Onychomadesis and palmoplantar keratoderma associated with talquetamab therapy for relapsed and refractory multiple myeloma.
- Author
-
Narayan N, Williams B, Lipe B, and De Benedetto A
- Abstract
Competing Interests: None disclosed.
- Published
- 2022
- Full Text
- View/download PDF
14. Selinexor-based regimens in patients with multiple myeloma after prior anti-B-cell maturation antigen treatment.
- Author
-
Baljevic M, Gasparetto C, Schiller GJ, Tuchman SA, Callander NS, Lentzsch S, Monge J, Kotb R, Bahlis NJ, White D, Chen CI, Sutherland HJ, Madan S, LeBlanc R, Sebag M, Venner CP, Bensinger WI, Biran N, DeCastro A, Van Domelen DR, Zhang C, Shah JJ, Shacham S, Kauffman MG, Bentur OS, and Lipe B
- Abstract
There is a lack of consensus on therapy sequencing in previously treated multiple myeloma, particularly after anti-B-cell maturation antigen (BCMA) therapy. Earlier reports on selinexor (X) regimens demonstrated considerable efficacy in early treatment, and after anti-BCMA-targeted chimeric antigen receptor-T cell therapy. Here, we present data from 11 heavily pretreated patients who predominantly received BCMA-antibody-drug conjugate therapy. We observe that X-containing regimens are potent and achieve durable responses with numerically higher overall response and clinical benefit rates, as well as median progression free survival compared to patients' prior anti-BCMA therapies, despite being used later in the treatment course. In an area of evolving unmet need, these data reaffirm the efficacy of X-based regimens following broader anti-BCMA therapy., Competing Interests: Muhamed Baljevic ‐ Consultancy: BMS/Celgene, Cardinal Health; Sanofi‐Genzyme. Advisory Boards: Oncopeptides, Janssen Research, Karyopharm, BMS/Celgene, Sanofi‐Genzyme. Speaker: NCCN, CurioScience, AJH. Cristina Gasparetto ‐ Leadership: Celgene. Consulting or Advisory Role: Abbvie/Genentech; Celgene; GlaxoSmithKline; Janssen; Karyopharm Therapeutics; Sanofi. Speaker: GlaxoSmithKline; Karyopharm Therapeutics; Sanofi. Travel, Accommodations, Expenses: Celgene; Karyopharm Therapeutics. Gary J. Schiller ‐ clinical research support from KaryopharmSascha A. Tuchman ‐ Consulting: Caelum, Sanofi, Shattuck Labs, Janssen; Research support: Karyopharm, Sanofi, Caelum, Janssen. Natalie S. Callander ‐ Research Funding: Cellectar. Suzanne Lentzsch ‐ Leadership: Caelum Biosciences. Stock and Other Ownership Interests: Caelum Biosciences. Consulting or Advisory Role: Abbvie/Genentech; Amgen; Caelum Biosciences; Celularity; GlaxoSmithKline; Janssen; Sanofi; Sorrento Therapeutics; Takeda. Speaker: Clinical Care Options/NCCN; PeerView. Research Funding: Karyopharm Therapeutics; Sanofi. Patents, Royalties, Other Intellectual Property: Patent 11‐1F4 mAb for use in AL Amyloidosis. Travel, Accommodations, Expenses: Janssen. Jorge Monge ‐ Consultancy for BMS, Research funding from Karyopharm Therapeutics. Rami Kotb ‐ Research funding: Merck, Sanofi. Ownership/Share holder: Karyopharm. Honoraria: Celgene/BMS, Janssen, Takeda, Amgen, Sanofi, Merck, Pfizer. Nizar J. Bahlis ‐ Consultancy and advisory board: BMS/Celgene, Janssen, Pfizer, Amgen, Genentech, Sanofi, Karyopharm. Research funding: PfizerDarrell White‐ Amgen, Antengene, Celgene/BMS, Forus, GSK, Janssen, Karyopharm, Sanofi, Takeda: honoraria, consultancyChristine I. Chen ‐ Consulting or Advisory Role ‐ Abbvie; AstraZeneca; Bristol‐Myers Squibb; Gilead Sciences; Janssen; Novartis; Research Funding ‐ Gilead Sciences. Heather J. Sutherland ‐ Honoraria ‐ Amgen; Bristol‐Myers Squibb; Celgene; Genzyme; Janssen; Takeda. Consulting or advisory role ‐ Amgen; Bristol‐Myers Squibb; Celgene; Janssen; Sanofi; Takeda. Sumit Madan ‐ Speaker bureau: Janssen, BMS Ad hoc advisory board/consultancy: Janssen, Takeda, Oncopeptide, Pfizer. Richard LeBlanc‐ Consultancy/advisory board: BMS Canada; Janssen Inc.; Amgen Canada; Takeda Canada; Sanofi Canada. Michael Sebag ‐ Honoraria: Amgen; Bristol‐Myers Squibb; Celgene; Janssen‐Ortho; Karyopharm Therapeutics; Novartis; Takeda. Research funding: Janssen. Patents, Royalties, Other. Intellectual Property: Patent but with no associated royalties or profit. Christopher P. Venner ‐ Honoraria: Amgen; Bristol‐Myers Squibb; GlaxoSmithKline; Janssen; Sanofi; Takeda. William I. Bensinger ‐ Speakers bureau: Janssen, BMS, Amgen, Takeda, SanofiDane R. Van Domelen, Ohad S. Bentur, and Chris Zhang are employees of Karyopharm Therapeutics. Andrew DeCastro, Jatin J. Shah, and Michael G. Kauffman are former employees of Karyopharm Therapeutics. Sharon Shacham is a former employee of Karyopharm Therapeutics and holds patents (8999996, 9079865, 9714226, PCT/US12/048319, and I574957) on hydrazide‐containing nuclear transport modulators and uses and holding pending patents (PCT/US12/048319, 499/2012, PI20102724, and 2012000928) on hydrazide‐containing nuclear transport modulators and uses. Brea Lipe ‐ Consultant for BMS, Janssen, GSK., (© 2022 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
15. Laboratory Detection and Initial Diagnosis of Monoclonal Gammopathies.
- Author
-
Keren DF, Bocsi G, Billman BL, Etzell J, Faix JD, Kumar S, Lipe B, McCudden C, Montgomery R, Murray DL, Rai AJ, Redondo TC, Souter L, Ventura CB, and Ansari MQ
- Subjects
- Humans, Laboratories, Systematic Reviews as Topic, Paraproteinemias diagnosis
- Abstract
Context.—: The process for identifying patients with monoclonal gammopathies is complex. Initial detection of a monoclonal immunoglobulin protein (M protein) in the serum or urine often requires compilation of analytical data from several areas of the laboratory. The detection of M proteins depends on adequacy of the sample provided, available clinical information, and the laboratory tests used., Objective.—: To develop an evidence-based guideline for the initial laboratory detection of M proteins., Design.—: To develop evidence-based recommendations, the College of American Pathologists convened a panel of experts in the diagnosis and treatment of monoclonal gammopathies and the laboratory procedures used for the initial detection of M proteins. The panel conducted a systematic literature review to address key questions. Using the Grading of Recommendations Assessment, Development, and Evaluation approach, recommendations were created based on the available evidence, strength of that evidence, and key judgements as defined in the Grading of Recommendations Assessment, Development, and Evaluation Evidence to Decision framework., Results.—: Nine guideline statements were established to optimize sample selection and testing for the initial detection and quantitative measurement of M proteins used to diagnose monoclonal gammopathies., Conclusions.—: This guideline was constructed to harmonize and strengthen the initial detection of an M protein in patients displaying symptoms or laboratory features of a monoclonal gammopathy. It endorses more comprehensive initial testing when there is suspicion of amyloid light chain amyloidosis or neuropathies, such as POEMS (polyneuropathy, organomegaly, endocrinopathy, M protein, and skin changes) syndrome, associated with an M protein., Competing Interests: Authors' disclosures of potential conflicts of interest and author contributions are found in the Appendix at the end of this article.
- Published
- 2022
- Full Text
- View/download PDF
16. SOHO State of the Art Updates and Next Questions: Treatment of Older, Vulnerable Adults with Multiple Myeloma.
- Author
-
Grant SJ, Joshi G, and Lipe B
- Subjects
- Aged, Humans, Transplantation, Autologous, Frailty, Hematopoietic Stem Cell Transplantation, Multiple Myeloma drug therapy, Multiple Myeloma therapy
- Abstract
Multiple myeloma is primarily a disease of the elderly, and optimal treatments must weigh the risks of toxicity with the benefits of therapy. Frailty scales have been developed to aid treatment-decision making for older adults with MM. This review provides a framework for incorporating frailty scales into clinical care and highlights how patient-aligned priorities for care can influence the management of older or more vulnerable adults with newly diagnosed multiple myeloma newly diagnosed multiple myeloma. We review the currently available systemic therapies for managing older or more vulnerable adults with newly diagnosed multiple myeloma otherwise considered ineligible for autologous stem cell transplantation., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2022
- Full Text
- View/download PDF
17. Cell-free DNA for the detection of emerging treatment failure in relapsed/ refractory multiple myeloma.
- Author
-
Waldschmidt JM, Yee AJ, Vijaykumar T, Pinto RA, Frede J, Anand P, Bianchi G, Guo G, Potdar S, Seifer C, Nair MS, Kokkalis A, Kloeber JA, Shapiro S, Budano L, Mann M, Friedman R, Lipe B, Campagnaro E, O'Donnell EK, Zhang CZ, Laubach JP, Munshi NC, Richardson PG, Anderson KC, Raje NS, Knoechel B, and Lohr JG
- Subjects
- Humans, Treatment Failure, Cell-Free Nucleic Acids genetics, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, Multiple Myeloma genetics
- Abstract
Interrogation of cell-free DNA (cfDNA) represents an emerging approach to non-invasively estimate disease burden in multiple myeloma (MM). Here, we examined low-pass whole genome sequencing (LPWGS) of cfDNA for its predictive value in relapsed/ refractory MM (RRMM). We observed that cfDNA positivity, defined as ≥10% tumor fraction by LPWGS, was associated with significantly shorter progression-free survival (PFS) in an exploratory test cohort of 16 patients who were actively treated on diverse regimens. We prospectively determined the predictive value of cfDNA in 86 samples from 45 RRMM patients treated with elotuzumab, pomalidomide, bortezomib, and dexamethasone in a phase II clinical trial (NCT02718833). PFS in patients with tumor-positive and -negative cfDNA after two cycles of treatment was 1.6 and 17.6 months, respectively (HR 7.6, P < 0.0001). Multivariate hazard modelling confirmed cfDNA as independent risk factor (HR 96.6, P = 6.92e-05). While correlating with serum-free light chains and bone marrow, cfDNA additionally discriminated patients with poor PFS among those with the same response by IMWG criteria. In summary, detectability of MM-derived cfDNA, as a measure of substantial tumor burden with therapy, independently predicts poor PFS and may provide refinement for standard-of-care response parameters to identify patients with poor response to treatment earlier than is currently feasible., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
18. Once weekly selinexor, carfilzomib and dexamethasone in carfilzomib non-refractory multiple myeloma patients.
- Author
-
Gasparetto C, Schiller GJ, Tuchman SA, Callander NS, Baljevic M, Lentzsch S, Rossi AC, Kotb R, White D, Bahlis NJ, Chen CI, Sutherland HJ, Madan S, LeBlanc R, Sebag M, Venner CP, Bensinger WI, Biran N, Ammu S, Ben-Shahar O, DeCastro A, Van Domelen D, Zhou T, Zhang C, Bentur OS, Shah J, Shacham S, Kauffman M, and Lipe B
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Dexamethasone adverse effects, Drug Administration Schedule, Female, Humans, Hydrazines adverse effects, Male, Maximum Tolerated Dose, Middle Aged, Multiple Myeloma genetics, Oligopeptides adverse effects, Survival Analysis, Translocation, Genetic, Treatment Outcome, Triazoles adverse effects, Dexamethasone administration & dosage, Hydrazines administration & dosage, Multiple Myeloma drug therapy, Oligopeptides administration & dosage, Triazoles administration & dosage
- Abstract
Background: Proteasome inhibitors (PIs), including carfilzomib, potentiate the activity of selinexor, a novel, first-in-class, oral selective inhibitor of nuclear export (SINE) compound, in preclinical models of multiple myeloma (MM)., Methods: The safety, efficacy, maximum-tolerated dose (MTD) and recommended phase 2 dose (RP2D) of selinexor (80 or 100 mg) + carfilzomib (56 or 70 mg/m
2 ) + dexamethasone (40 mg) (XKd) once weekly (QW) was evaluated in patients with relapsed refractory MM (RRMM) not refractory to carfilzomib., Results: Thirty-two patients, median prior therapies 4 (range, 1-8), were enrolled. MM was triple-class refractory in 38% of patients and 53% of patients had high-risk cytogenetics del(17p), t(4;14), t(14;16) and/or gain 1q. Common treatment-related adverse events (all/Grade 3) were thrombocytopenia 72%/47% (G3 and G4), nausea 72%/6%, anaemia 53%/19% and fatigue 53%/9%, all expected and manageable with supportive care and dose modifications. MTD and RP2D were identified as selinexor 80 mg, carfilzomib 56 mg/m2 , and dexamethasone 40 mg, all QW. The overall response rate was 78% including 14 (44%) ≥ very good partial responses. Median progression-free survival was 15 months., Conclusions: Weekly XKd is highly effective and well-tolerated. These data support further investigation of XKd in patients with MM., (© 2021. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
19. Consensus guidelines and recommendations for infection prevention in multiple myeloma: a report from the International Myeloma Working Group.
- Author
-
Raje NS, Anaissie E, Kumar SK, Lonial S, Martin T, Gertz MA, Krishnan A, Hari P, Ludwig H, O'Donnell E, Yee A, Kaufman JL, Cohen AD, Garderet L, Wechalekar AF, Terpos E, Khatry N, Niesvizky R, Yi Q, Joshua DE, Saikia T, Leung N, Engelhardt M, Mothy M, Branagan A, Chari A, Reiman AJ, Lipe B, Richter J, Rajkumar SV, Miguel JS, Anderson KC, Stadtmauer EA, Prabhala RH, McCarthy PL, and Munshi NC
- Subjects
- Consensus, Humans, Infections complications, Multiple Myeloma complications, Multiple Myeloma drug therapy
- Abstract
Infection remains the leading cause of morbidity and mortality in patients with multiple myeloma because of the cumulative effect of disease, treatment, and host-related factors. Given that infectious risk is cumulative through the course of the disease, preventing infections is paramount. Optimal preventive strategies include vaccination against common pathogens, antimicrobial prophylaxis, infection control measures, and immunoglobulin replacement in a small subset of patients; however, there are no universally accepted guidelines for infection prevention. This Review provides a consensus statement from a panel of 36 experts with global representation, which was convened by The International Myeloma Society to review existing literature and current guidelines, address issues associated with the risk of infection and prevention of infectious complications in multiple myeloma in the context of emerging therapies, and offer recommendations for preventing these complications., Competing Interests: Declaration of interests NSR reports grants and consulting fees from Bristol Myers Squibb–Celgene, Bluebird Bio, Amgen, Janssen, GlaxoSmithKline, Caribou Biosciences, and Immuneel; and personal fees from Research to Practice, Karyopharm, Takeda, Amgen, and Janssen, outside the submitted work. SKK reports grants and consulting fees from Bristol Myers Squibb–Celgene, Takeda, Abbvie, Roche, and Janssen; grants from Medimmune, Tenebio, and Carsgen; and personal fees from Oncopeptides, Beigene, and Antengene, outside the submitted work. SL reports personal fees from Celgene, Janssen, Takeda, Novartis, Bristol Myers Squibb, GlaxoSmithKline, and Amgen, outside the submitted work. TM reports personal fees from GlaxoSmithKline; and grants from Sanofi, Janssen, and Amgen, outside the submitted work. MAG reports personal fees from Abbvie, Celgene, Akcea, i3Health, Prothena, Research to Practice, Alnylym, Ambry Genetric, Amgen, Janssen, Celgene, Aurora Bio, Ionis, Karyopharm, and Sanofi; and grants from Pfizer, outside the submitted work. PH reports grants and personal fees from Bristol Myers Squibb, Takeda, Amgen, Janssen, and Legend Biotech, outside the submitted work. HL reports personal fees from Janssen, Bristol Myers Squibb–Celgene, and Seattle Genetics; and grants and personal fees from Amgen and Takeda, outside the submitted work. JLK reports grants from Abbvie; personal fees from TG Therapeutics and Incyte; and grants and personal fees from Janssen, Bristol Myers Squibb, Amgen, and Takeda, outside the submitted work. ADC reports personal fees from Bristol Myers Squibb–Celgene, Janssen, Takeda, AstraZeneca, Genentech–Roche, Oncopeptides, and Seattle Genetics; grants from Novartis; and grants and personal fees from GlaxoSmithKline, outside the submitted work. LG reports personal fees from Amgen, Takeda, Novartis, Bristol Myers Squibb, Janssen, and Sanofi, outside the submitted work. ET reports personal fees from Bristol Myers Squibb; grants and personal fees from GlaxoSmithKline, Janssen, and Sanofi; and grants, personal fees, and consulting fees from Amgen, Genesis Pharma, and Takeda, outside the submitted work. RN reports personal fees from Takeda, Amgen, Celgene, Janssen, Karyopharm, GlaxoSmithKline, and Bristol Myers Squibb, outside the submitted work. NL reports consulting fees from AbbVie, Takeda, and Omeros; and grants from Omeros and Alnylam, outside the submitted work. ME reports grants from Amgen, Bristol Myers Squibb, Janssen, Takeda, Sanofi, and GlaxoSmithKline, outside the submitted work. AB reports personal fees from BeiGene, Sanofi Genzyme, Pharmacyclics, and Karyopharm, outside the submitted work. AC reports personal fees from Bristol Myers Squibb, Karyopharm, Sanofi, Oncopeptides, Antengene, GlaxoSmithKline, Secura Bio, and Shattuck Labs; grants from Pharmacyclics; and grants and personal fees from Janssen, Celgene, Novartis Pharmaceuticals, Amgen, Seattle Genetics, and Millenium–Takeda, outside the submitted work. BL reports personal fees from Karyopharm, Bristol Myers Squibb, Janssen, and GlaxoSmithKline; grants from Cellectar; and grants and personal fees from Amgen, outside the submitted work. JR reports personal fees from Bristol Myers Squibb, Takeda, Karyopharm, Oncopeptides, AstraZeneca, Adaptive Biotechnologies, Janssen, Secura Bio, and Sanofi, outside the submitted work. JSM reports personal fees from Amgen, Bristol Myers Squibb, Celgene, Janssen, Merck, Novartis, Takeda, Sanofi, Roche, GlaxoSmithKline, Abbvie, Karyopharm, and SecuraBio, outside the submitted work. KCA reports personal fees from Amgen, Pfizer, Janssen, AstraZeneca, Precision Biosciences, Windmill, Mana, Starton, Raqia, Oncopeptides, and C4 Therapeutics, outside the submitted work. PLM reports personal fees from BlueBird Biotech, Bristol Myers Squibb, Fate Therapeutics, Janssen, Juno, Karyopharm, Magenta Therapeutics, Takeda, and Medscape; grants from Celgene; and non-financial support from Sanofi, outside the submitted work. NCM reports personal fees from Bristol Myers Squibb, Oncopeptides, Janssen, Amgen, Novartis, Takeda, Abbvie, and C4 Therapeutics, outside the submitted work. All other authors declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
20. Bisphosphonates for delivering drugs to bone.
- Author
-
Sun S, Tao J, Sedghizadeh PP, Cherian P, Junka AF, Sodagar E, Xing L, Boeckman RK Jr, Srinivasan V, Yao Z, Boyce BF, Lipe B, Neighbors JD, Russell RGG, McKenna CE, and Ebetino FH
- Subjects
- Animals, Bacteria, Biofilms, Humans, Quality of Life, Diphosphonates, Pharmaceutical Preparations
- Abstract
Advances in the design of potential bone-selective drugs for the treatment of various bone-related diseases are creating exciting new directions for multiple unmet medical needs. For bone-related cancers, off-target/non-bone toxicities with current drugs represent a significant barrier to the quality of life of affected patients. For bone infections and osteomyelitis, bacterial biofilms on infected bones limit the efficacy of antibiotics because it is hard to access the bacteria with current approaches. Promising new experimental approaches to therapy, based on bone-targeting of drugs, have been used in animal models of these conditions and demonstrate improved efficacy and safety. The success of these drug-design strategies bodes well for the development of therapies with improved efficacy for the treatment of diseases affecting the skeleton. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc., (©2020 The British Pharmacological Society.)
- Published
- 2021
- Full Text
- View/download PDF
21. Bortezomib, lenalidomide, and dexamethasone with or without elotuzumab in patients with untreated, high-risk multiple myeloma (SWOG-1211): primary analysis of a randomised, phase 2 trial.
- Author
-
Usmani SZ, Hoering A, Ailawadhi S, Sexton R, Lipe B, Hita SF, Valent J, Rosenzweig M, Zonder JA, Dhodapkar M, Callander N, Zimmerman T, Voorhees PM, Durie B, Rajkumar SV, Richardson PG, and Orlowski RZ
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bortezomib administration & dosage, Bortezomib adverse effects, Dexamethasone administration & dosage, Dexamethasone adverse effects, Disease-Free Survival, Female, Humans, Lenalidomide administration & dosage, Lenalidomide adverse effects, Maintenance Chemotherapy, Male, Middle Aged, Survival Rate, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Multiple Myeloma drug therapy, Multiple Myeloma genetics, Multiple Myeloma metabolism
- Abstract
Background: The introduction of immunomodulatory agents, proteasome inhibitors, and autologous haematopoietic stem-cell transplantation has improved outcomes for patients with multiple myeloma, but patients with high-risk multiple myeloma have a poor long-term prognosis. We aimed to address optimal treatment for these patients., Methods: SWOG-1211 is a randomised phase 2 trial comparing eight cycles of lenalidomide (25 mg orally on days 1-14 every 21 days), bortezomib (1·3 mg/m
2 subcutaneously on days 1, 4, 8, and 11 every 21 days), and dexamethasone (20 mg orally on days 1, 2, 4, 5, 8, 9, 11, and 12 every 21 days; RVd) induction followed by dose-attenuated RVd maintenance (bortezomib 1 mg/m2 subcutaneously on days 1, 8, and 15; lenalidomide 15 mg orally on days 1-21; dexamethasone 12 mg orally on days 1, 18, and 15 every 28 days) until disease progression with or without elotuzumab (10 mg/kg intravenously on days 1, 8, and 15 for cycles 1-2, on days 1 and 11 for cycles 3-8, and on days 1 and 15 during maintenance). Patients were randomly assigned (1:1) to either RVd or RVd-elotuzumab. High-risk multiple myeloma was defined by one of the following: gene expression profiling high risk (GEPhi ), t(14;16), t(14;20), del(17p) or amp1q21, primary plasma cell leukaemia and elevated serum lactate dehydrogenase (two times the upper limit of normal or more). The primary endpoint was progression-free survival, and all analyses were done on intention-to-treat basis among eligible patients who were evaluable for response. This study is registered with ClinicalTrials.gov, NCT01668719., Findings: 100 (RVd n=52, RVd-elotuzumab n=48) patients were enrolled between Oct 27, 2013, and May 15, 2016, across 26 cooperative group institutions in the USA. Median age was 64 years (IQR 57-70, range 36-85). 74 (75%) of 99 had International Staging System stage II or stage III disease, 47 (47%) of 99 had amp1q21, 37 (37%) of 100 had del17p, 11 (11%) of 100 had t(14;16), eight (9%) of 90 were GEPhi , seven (7%) of 100 had primary plasma cell leukaemia, five (5%) of 100 had t(14;20), four (4%) of 100 had elevated serum lactate dehydrogenase, and 17 (17%) had two or more features. With a median follow-up of 53 months (IQR 46-59), no difference in median progression-free survival was observed (RVd 33·64 months [95% CI 19·55-not reached], RVd-elotuzumab 31·47 months [18·56-53·98]; hazard ratio 0·968 [80% CI 0·697-1·344]; one-sided p=0·45]. 37 (71%) of 52 patients in the RVd group and 37 (77%) of 48 in the RVd-elotuzumab group had grade 3 or worse adverse events. No significant differences in the safety profile were observed, although some notable results included grade 3-5 infections (four [8%] of 52 in the RVd group, eight [17%] of 48 in the RVd-elotuzumab group), sensory neuropathy (four [8%] of 52 in the RVd group, six [13%] of 48 in the RVd-elotuzumab group), and motor neuropathy (one [2%] of 52 in the RVd group, four [8%] of 48 in the RVd-elotuzumab group). There were no treatment-related deaths in the RVd group and one death in the RVd-elotuzumab group for which study treatment was listed as possibly contributing by the investigator., Interpretation: In the first randomised study of high-risk multiple myeloma reported to date, the addition of elotuzumab to RVd induction and maintenance did not improve patient outcomes. However, progression-free survival in both study groups exceeded the original statistical assumptions and supports the role for continuous proteasome inhibitors and immunomodulatory drug combination maintenance therapy for this patient population., Funding: National Institutes of Health, National Cancer Institute, Bristol Myers Squibb, Celgene, Leukemia and Lymphoma Society., (Copyright © 2021 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
22. Selinexor, daratumumab, and dexamethasone in patients with relapsed or refractory multiple myeloma.
- Author
-
Gasparetto C, Lentzsch S, Schiller G, Callander N, Tuchman S, Chen C, White D, Kotb R, Sutherland H, Sebag M, Baljevic M, Bensinger W, LeBlanc R, Venner C, Bahlis N, Rossi A, Biran N, Sheehan H, Saint-Martin JR, Van Domelen D, Kai K, Shah J, Shacham S, Kauffman M, and Lipe B
- Abstract
We assessed the safety, efficacy, maximum tolerated dose (MTD), and the recommended phase 2 dose (RP2D) of selinexor, a first in class oral selective inhibitor of nuclear export (100 mg once weekly [QW] or 60 mg twice weekly), in combination with daratumumab (16 mg/kg per label) and dexamethasone (40 mg QW) (SDd) in patients with relapsed refractory multiple myeloma (RRMM). Thirty-four patients (median prior therapies, 3 [range, 2-10]) were enrolled; MM was refractory to proteasome inhibitor (PI) in 85%, immunomodulatory agent (IMiD) in 76%, both in 74%, and daratumumab in 6% of patients. Two dose-limiting toxicities (DLTs) were reported in the selinexor 60 mg twice-weekly cohort with no DLTs in the 100 mg QW cohort, making 100 mg QW the MTD and RP2D. Common treatment-related adverse events included thrombocytopenia (70.6%), nausea (70.6%), fatigue (61.8%), anemia (61.8%), and neutropenia (50.0%). Overall response rate was 73% and median progression-free survival 12.5 months in daratumumab-naïve patients. SDd was well tolerated and its promising efficacy suggests that further study of this PI- and IMiD-free regimen in RRMM patients who had at least one prior line of therapy including a PI and an IMiD but whose disease is naïve to daratumumab is warranted., Competing Interests: Cristina Gasparetto: No conflict of interest; Suzanne Lentzsch: research funding—Karyopharm and Sanofi; patents, royalties, other intellectual property—Caelum Bioscience; stock and other ownership interests—Caelum Bioscience, Mesoblast, Magenta, and Kadmon; consulting or advisory role—Caelum Bioscience, Sorrento, Janssen, and Celularity; Gary Schiller: research funding: AbbVie, Agios, Actinium, Ambit, AMGEN, ARIAD, Astellas, Leukemia & Lymphoma Society, BioMed Valley Discoveries, Inc., Bluebird Bio, Bristol‐Myers Squibb, Boehringer‐Ingleheim, Celator, Celgene, Cellerant, Constellation Pharmaceuticals, CTI BioPharma Corp., Forma, Cyclacel, Daiichi Sankyo, Deciphera, The California Institute for Regenerative Medicine (CIRM), Gamida Cell Ltd., GILEAD, Incyte, Janssen, Karyopharm, Kite Pharma, Inc., Mateon, MedImmune, Millennium, National Marrow Donor Program, National Institute of Health: National Cancer Institute, Novartis, Onconova, Onyx, Pfizer, PharmaMar, Sangamo, Stemline Therapeutics, Inc., National Marrow Donor Program, Tolero, Trovagene, University of California Davis, and University of California San Diego‐ UCHMC; stock and other ownership interests—Amgen, Bristol‐Myers Squibb, Pfizer, and Johnson and Johnson; consulting or advisory role—Incyte, Elevate Bio, AbbVie, ONO UK, Novartis, Evidera, Agios, AstraZeneca, National Institute of Health: National Cancer Institute, and Federal Drug Administration; speakers' bureau—Agios, Amgen, Astellas, Bristol‐Myers Squibb, Celgene, Sanofi‐Genzyme, Incyte, Janssen, Jazz, Kite (gilead)‐Yescarta, Pharmacyclics, and Stemline; Natalie Callander: research funding—Cellectar; Sascha Tuchman: research funding—Celgene, Karyopharm, Amgen, Janssen, and Sanofi; consulting or advisory role—Oncopeptides, Celgene, Karyopharm, Caelum, and Sanofi; honoraria—Celgene, Karyopharm, Caelum, and Sanofi; speakers' bureau—Celgene; Christine Chen: No conflict of interest; Darrell White: consulting or advisory role: Amgen, Antengene, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Karyopharm, Sanofi, and Takeda; honoraria—Amgen, Antengene, Bristol Myers Squibb, Celgene, GlaxoSmithKline, Janssen, Karyopharm, Sanofi, and Takeda; Rami Kotb: research funding—Merck and Sanofi; stock and other ownership interests—Karyopharm; consulting or advisory role—Celgene/BMS, Janssen, Amgen, Takeda, Sanofi, and Merck; Heather Sutherland: honoraria: Amgen, Celgene, Janssen, Takeda, Bristol Myers Squibb, and GlaxoSmith Kline; Michael Sebag: consulting or advisory role—Janssen and Karyopharm; Muhamed Baljevic: consulting or advisory role—Celgene Corporation, Cardinal Health, Putnam Associates, Gerson Lehrman Group, Inc., and AlphaSights; honoraria—Karyopharm Therapeutics Inc. clinical trial internal review committee member and NCCN Hematologic Malignancies Congress panelist; William Bensinger: research funding—BMS, Acetylon, Amgen, Janssen, Regeneron, and Sanofi; consulting or advisory role—Regeneron and BMS; speakers' bureau—Amgen, Janssen, BMS, Sanofi, and GSK; travel, accommodations, and expenses: Amgen, Janssen, BMS, Sanofi, and GSK; Richard LeBlanc: consulting or advisory role—Celgene Canada, Janssen Inc., Amgen Canada, Takeda Canada, Sanofi Canada; speakers' bureau—Celgene Canada, Janssen Inc., and Amgen Canada; Chris Venner: honoraria—Celgene, Johnson & Johnson, Amgen, Sanofi, and Takeda; Nizar Bahlis: research funding—received research support from Celegen and BMS; honoraria—Janssen, Celgene/BMS. Amgen, Takeda, Karyopharm, Sanofi, and GSK; Heidi Sheehan: employee and stockholder in Karyopharm Therapeutics; Jean‐Richard Saint‐Martin: employee and stockholder in Karyopharm Therapeutics; Dane Van Domelen: employee and stockholder in Karyopharm Therapeutics; Kazuharu Kai: employee and stockholder in Karyopharm Therapeutics; Jatin Shah: Executive Vice President, CMO, and stockholder in Karyopharm Therapeutics; Sharon Shacham: President, CSO, and stockholder in Karyopharm Therapeutics; Michael Kauffman: CEO and stockholder in Karyopharm Therapeutics; Brea Lipe: research funding—Amgen and Cellectar; consulting or advisory role—BMS, Janssen, and Abbvie., (© 2020 The Authors. eJHaem published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
23. Management of Frail Older Adults with Newly Diagnosed Multiple Myeloma - Moving Toward a Personalized Approach.
- Author
-
Grant SJ and Lipe B
- Subjects
- Age Factors, Aged, Frail Elderly, Humans, Male, Multiple Myeloma therapy, Precision Medicine methods
- Abstract
Case Vignette: A 75-year-old male undergoes an evaluation for progressively worsening fatigue with associated shortness of breath. He also reports back pain worse at night, which does not resolve with acetaminophen. He is retired, but over the past 2 months he is unable to garden or perform household chores. Due to his pain and fatigue, he has limited his activity. He currently takes 9 prescription medications for chronic medical conditions, which include diabetes mellitus with neuropathy, hypertension, hyperlipidemia, and COPD. Initial evaluation reveals anemia (Hgb 9.0 g/dl last known normal was 2 years ago, Hgb 13.5 g/dl), renal impairment (serum creatinine 2.5 mg/dL), hypercalcemia (11.5 g/dl). Plain X-rays reveal compression fractures involving T3 and T4. Multiple myeloma is suspected, further labs confirm the diagnosis of IgG kappa multiple myeloma. LDH is elevated, beta-2-microglobulin is elevated at 6.1 mg/L and albumin < 3.5 mg/dl. Bone marrow aspiration and biopsy reveal plasmacytosis of 55% and on fluorescence in situ hybridization testing, del 17p/TP53 mutation in 85% of cells is detected. PET/CT confirms diffuse bone disease involving the axial and appendicular skeleton. His Karnofsky performance status (KPS) is 70%. He is widowed and lives alone but has 2 adult children who currently live out of state. This abstract will discuss how assessments of fitness/frailty may be used to develop personalized care tailored to the unique needs of older frail adults with multiple myeloma., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. Integrating Touchscreen-Based Geriatric Assessment and Frailty Screening for Adults With Multiple Myeloma to Drive Personalized Treatment Decisions.
- Author
-
Nathwani N, Kurtin SE, Lipe B, Mohile SG, Catamero DD, Wujcik D, Birchard K, Davis A, Dudley W, Stricker CT, and Wildes TM
- Subjects
- Aged, Aged, 80 and over, Female, Frail Elderly, Humans, Male, Mass Screening, Multiple Myeloma pathology, Pilot Projects, Prospective Studies, Geriatric Assessment methods, Multiple Myeloma diagnosis, Precision Medicine methods
- Abstract
Purpose: Geriatric assessment (GA) results predict toxicity/survival in older adults, yet GA is not routinely used in care for patients with multiple myeloma (MM). We tested a tablet-based modified GA (mGA) providing real-time results to clinicians., Methods: One hundred sixty-five patients with MM aged ≥ 65 years facing a treatment decision from 4 sites completed a tablet-based mGA with Katz Activities of Daily Living (ADL), Lawton Instrumental ADL, Charlson Comorbidity Index, and variables from the Cancer and Aging Research Group's Chemotherapy Toxicity Calculator. Providers reviewed the assessment results at the treatment visit., Results: Patients were white (72%; n = 86), mean age was 72 years (range, 65-85 years), and averaged 7.71 minutes (range, 2-17 minutes) for survey completion. Providers averaged 3.2 minutes (range, 1-10 minutes) to review mGA results. Using International Myeloma Working Group frailty score, patients were fit (39%; n = 64), intermediate fit (33%; n = 55), or frail (28%; n = 46). Providers selected more aggressive treatments in 16.3% of patients and decreased treatment intensity in 34% of patients; treatment intensification was more common for fit patients and milder treatments for frail patients (χ
2 = 20.02; P < .0001). Transplant eligibility significantly correlated with fit status and transplant ineligibility with frail status ( P = .004). Outcomes on 144 patients 3 months post study visit showed 19.4% (n = 28) had grade ≥ 3 hematologic toxicities, 38.9% (n = 56) had dose modifications, and 18% (n = 26) had early therapy cessation., Conclusion: Limited patient time required for survey completion and provider time for results review show mGA can be easily incorporated into clinical workflow. Real-time mGA results indicating fit/frailty status influenced treatment decisions.- Published
- 2020
- Full Text
- View/download PDF
25. A review of thrombotic microangiopathies in multiple myeloma.
- Author
-
Portuguese AJ, Gleber C, Passero FC Jr, and Lipe B
- Subjects
- Biomarkers, Complement Activation immunology, Complement System Proteins immunology, Disease Management, Humans, Multiple Myeloma drug therapy, Multiple Myeloma metabolism, Proteasome Inhibitors adverse effects, Proteasome Inhibitors therapeutic use, Research, Risk Factors, Thrombotic Microangiopathies therapy, Treatment Outcome, von Willebrand Factor metabolism, Multiple Myeloma complications, Thrombotic Microangiopathies diagnosis, Thrombotic Microangiopathies etiology
- Abstract
Patients with multiple myeloma (MM) are susceptible to developing thrombotic microangiopathies (TMAs), an etiologically diverse group of syndromes which include atypical hemolytic uremic syndrome (aHUS) and thrombotic thrombocytopenic purpura (TTP). The TMAs are characterized by thrombocytopenia and microangiopathic hemolytic anemia (MAHA), and are associated with a high mortality risk and irreversible end-organ damage when treatment is delayed. In MM patients, TMAs may be triggered by specific chemotherapies, bone marrow transplantation (BMT), and progression of underlying disease. Because many characteristics of TMAs overlap with sequelae of MM and its treatments, diagnosis requires a high index of suspicion. Furthermore, our understanding of optimal treatments for these entities is rapidly evolving and clinical practice guidelines do not yet exist. Historically, consideration of a diagnosis of TMA has prompted initiation of therapeutic plasma exchange. In this review, we present an overview of the MM-related TMAs, an approach to workup and diagnosis, and argue for initial empiric MM-related TMA treatment with eculizumab rather than plasma exchange., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Results of the First Clinical Study in Humans That Combines Hyperbaric Oxygen Pretreatment with Autologous Peripheral Blood Stem Cell Transplantation.
- Author
-
Abdelhakim H, Shune L, Bhatti S, Cantilena AR, Baran A, Lin TL, Ganguly S, Singh AK, Abhyankar S, Divine C, Lipe B, McGuirk J, Allin D, and Aljitawi OS
- Subjects
- Aged, Allografts, Disease-Free Survival, Female, Hematologic Neoplasms blood, Hematologic Neoplasms mortality, Humans, Male, Middle Aged, Pilot Projects, Survival Rate, Hematologic Neoplasms therapy, Hyperbaric Oxygenation, Peripheral Blood Stem Cell Transplantation
- Abstract
Patients undergoing high-dose chemotherapy and autologous hematopoietic cell transplantation (auto-HCT) are at risk for multiple morbidities, including mucosal inflammation and neutropenic fever, both related to neutropenia. Evidence from our preclinical work in an umbilical cord blood (UCB) transplantation murine model suggests that treatment with hyperbaric oxygen (HBO) before UCB infusion improves UCB CD34
+ cell engraftment by reducing erythropoietin levels. A pilot clinical trial using HBO in patients undergoing UCB transplantation showed improvement in kinetics of blood count recovery. In this study, we evaluated HBO in combination with auto-HCT. Our primary aim was to determine the safety of HBO in this setting and secondarily to determine its efficacy in reducing time to neutrophil and platelet engraftment compared with matched historic controls. Patients with multiple myeloma, non-Hodgkin lymphoma, and Hodgkin disease eligible for auto-HCT were included. On day 0, patients received HBO treatment consisting of exposure to 2.5 atmosphere absolutes for a total of 90 minutes, in a monoplace hyperbaric chamber, breathing 100% oxygen. Six hours after the start of HBO, peripherally mobilized stem/progenitor cells were infused and patients were followed daily for toxicity and blood count recovery. All patients received daily granulocyte colony-stimulating factor starting on day +5 and until absolute neutrophil count (ANC) of ≥1500 or ANC of 500 for 3 consecutive days. A matched historic cohort of 225 patients who received auto-HCT between January 2008 and December 2012 was chosen for comparison and matched on sex, age, conditioning regimen, and disease type. We screened 26 patients for this study; 20 were treated and included in the primary analysis, and 19 completed the HBO therapy and were included in the secondary analysis. Although the median time to neutrophil count recovery was 11 days in both the HBO and control cohorts, the Kaplan-Meier estimates of the full distributions indicate that the time to neutrophil recovery was generally about 1 day sooner for HBO versus historical controls (log-rank P = .005; range, 9 to 13 for HBO patients and 7 to 18 for controls). The median time to platelet count recovery was 16 days (range, 14 to 21) for HBO versus 18 days (range, 11 to 86) for controls (log-rank P < .0001). In the secondary analysis comparing the HBO cohort who completed HBO therapy (n = 19) with our historical cohort, we evaluated neutropenic fever, growth factor use, mucositis, day +100 disease responses, and blood product use. HBO was associated with less growth factor use (median 6 days in HBO cohort versus median 8 days in controls, P < .0001). Packed RBC and platelet transfusion requirements were not statistically different between the 2 cohorts. Mucositis incidence was significantly lower in the HBO cohort (26.3% in HBO cohort versus 64.2% in controls, P = .002). HBO therapy appears to be well tolerated in the setting of high-dose therapy and auto-HCT. Prospective studies are needed to confirm potential benefits of HBO with respect to earlier blood count recovery, reduced mucositis, and growth factor use, and a cost-benefit analysis is warranted. © 2019 American Society for Blood and Marrow Transplantation., (Copyright © 2019 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
27. Treatment of Multiple Myeloma: ASCO and CCO Joint Clinical Practice Guideline.
- Author
-
Mikhael J, Ismaila N, Cheung MC, Costello C, Dhodapkar MV, Kumar S, Lacy M, Lipe B, Little RF, Nikonova A, Omel J, Peswani N, Prica A, Raje N, Seth R, Vesole DH, Walker I, Whitley A, Wildes TM, Wong SW, and Martin T
- Subjects
- Clinical Trials, Phase II as Topic, Humans, Medical Oncology methods, Randomized Controlled Trials as Topic, Systematic Reviews as Topic, Medical Oncology standards, Multiple Myeloma therapy
- Abstract
Purpose: To provide evidence-based recommendations on the treatment of multiple myeloma to practicing physicians and others., Methods: ASCO and Cancer Care Ontario convened an Expert Panel of medical oncology, surgery, radiation oncology, and advocacy experts to conduct a literature search, which included systematic reviews, meta-analyses, randomized controlled trials, and some phase II studies published from 2005 through 2018. Outcomes of interest included survival, progression-free survival, response rate, and quality of life. Expert Panel members used available evidence and informal consensus to develop evidence-based guideline recommendations., Results: The literature search identified 124 relevant studies to inform the evidence base for this guideline., Recommendations: Evidence-based recommendations were developed for patients with multiple myeloma who are transplantation eligible and those who are ineligible and for patients with relapsed or refractory disease.
- Published
- 2019
- Full Text
- View/download PDF
28. Decellularized Wharton jelly matrix: a biomimetic scaffold for ex vivo hematopoietic stem cell culture.
- Author
-
Li D, Chiu G, Lipe B, Hopkins RA, Lillis J, Ashton JM, Paul S, and Aljitawi OS
- Subjects
- Antigens, CD34 analysis, Cell Differentiation, Cell Proliferation, Fetal Blood cytology, Humans, Transendothelial and Transepithelial Migration, Biomimetics methods, Cell Culture Techniques methods, Hematopoietic Stem Cells cytology, Tissue Scaffolds chemistry, Wharton Jelly chemistry
- Abstract
Hematopoietic stem progenitor cells (HSPCs) reside in the bone marrow (BM) hematopoietic "niche," a special 3-dimensional (3D) microenvironment that regulates HSPC self-renewal and multipotency. In this study, we evaluated a novel 3D in vitro culture system that uses components of the BM hematopoietic niche to expand umbilical cord blood (UCB) CD34
+ cells. We developed this model using decellularized Wharton jelly matrix (DWJM) as an extracellular matrix (ECM) scaffold and human BM mesenchymal stromal cells (MSCs) as supporting niche cells. To assess the efficacy of this model in expanding CD34+ cells, we analyzed UCB CD34+ cells, following culture in DWJM, for proliferation, viability, self-renewal, multilineage differentiation, and transmigration capability. We found that DWJM significantly expanded UCB HSPC subset. It promoted UCB CD34+ cell quiescence, while maintaining their viability, differentiation potential with megakaryocytic differentiation bias, and clonogenic capacity. DWJM induced an increase in the frequency of c-kit+ cells, a population with enhanced self-renewal ability, and in CXCR4 expression in CD34+ cells, which enhanced their transmigration capability. The presence of BM MSCs in DWJM, however, impaired UCB CD34+ cell transmigration and suppressed CXCR4 expression. Transcriptome analysis indicated that DWJM upregulates a set of genes that are specifically involved in megakaryocytic differentiation, cell mobility, and BM homing. Collectively, our results indicate that the DWJM-based 3D culture system is a novel in vitro model that supports the proliferation of UCB CD34+ cells with enhanced transmigration potential, while maintaining their differentiation potential. Our findings shed light on the interplay between DWJM and BM MSCs in supporting the ex vivo culture of human UCB CD34+ cells for use in clinical transplantation., (© 2019 by The American Society of Hematology.)- Published
- 2019
- Full Text
- View/download PDF
29. Induction Therapy for Newly Diagnosed Multiple Myeloma.
- Author
-
Paul B, Lipe B, Ocio EM, and Usmani SZ
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Clinical Decision-Making, Europe, Humans, Molecular Targeted Therapy, Multiple Myeloma diagnosis, Multiple Myeloma mortality, Neoplasm Staging, Neoplasm, Residual, Prognosis, Remission Induction, United States, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Multiple Myeloma therapy
- Abstract
The frontline therapy for newly diagnosed multiple myeloma (MM) has continued to evolve over the last 10 years. There has been a growing emphasis on achieving the best depth of response in the context of minimal residual disease negativity, given its prognostic correlation with superior overall survival. Another important area of emphasis has been to improve prognostication and staging by including information on disease biology. There also a growing appreciation of global differences in drug access and patterns of care. The current review explores each of these areas and how best to incorporate the emerging induction regimens in to schema of MM therapy.
- Published
- 2019
- Full Text
- View/download PDF
30. Hemophagocytic Lymphohistiocytosis in a Patient With Multiple Myeloma.
- Author
-
Hsu CM, Bennett JM, and Lipe B
- Subjects
- Humans, Lymphohistiocytosis, Hemophagocytic pathology, Male, Middle Aged, Lymphohistiocytosis, Hemophagocytic etiology, Multiple Myeloma complications
- Published
- 2019
- Full Text
- View/download PDF
31. Carfilzomib-induced aHUS responds to early eculizumab and may be associated with heterozygous CFHR3-CFHR1 deletion.
- Author
-
Portuguese AJ and Lipe B
- Subjects
- ADAMTS13 Protein metabolism, Aged, Antineoplastic Agents therapeutic use, Blood Component Removal, Creatinine blood, Female, Hemolytic-Uremic Syndrome etiology, Hemolytic-Uremic Syndrome genetics, Humans, Male, Middle Aged, Oligopeptides therapeutic use, Platelet Count, Sequence Deletion, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents adverse effects, Blood Proteins genetics, Complement C3b Inactivator Proteins genetics, Hemolytic-Uremic Syndrome drug therapy, Oligopeptides adverse effects
- Published
- 2018
- Full Text
- View/download PDF
32. A novel extracellular matrix-based leukemia model supports leukemia cells with stem cell-like characteristics.
- Author
-
Li D, Lin TL, Lipe B, Hopkins RA, Shinogle H, and Aljitawi OS
- Subjects
- Cell Culture Techniques methods, Cell Differentiation, Extracellular Matrix metabolism, Extracellular Matrix pathology, Extracellular Matrix Proteins metabolism, HL-60 Cells, Humans, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells pathology, Wharton Jelly metabolism, Wharton Jelly pathology, Extracellular Matrix chemistry, Extracellular Matrix Proteins chemistry, Leukemia, Myeloid, Acute metabolism, Models, Biological, Neoplastic Stem Cells metabolism, Wharton Jelly chemistry
- Abstract
Acute myeloid leukemia (AML) relapse results from the survival of chemotherapy-resistant and quiescent leukemia stem cells (LSC). These LSCs reside in the bone marrow microenvironment, comprised of other cells and extracellular matrix (ECM), which facilitates LSC quiescence through expression of cell adhesion molecules. We used decellularized Wharton's jelly matrix (DWJM), the gelatinous material in the umbilical cord, as a scaffolding material to culture leukemia cells, because it contains many components of the bone marrow extracellular matrix, including collagen, fibronectin, lumican, and hyaluronic acid (HA). Leukemia cells cultured in DWJM demonstrated decreased proliferation without undergoing significant differentiation. After culture in DWJM, these cells also exhibited changes in morphology, acquiring a spindle-shaped appearance, and an increase in the ALDH
+ cell population. When treated with a high-dose of doxorubicin, leukemia cells in DWJM demonstrated less apoptosis compared with cells in suspension. Serial colony forming unit (CFU) assays indicated that leukemia cells cultured in DWJM showed increased colony-forming ability after both primary and secondary plating. Leukemia cell culture in DWJM was associated with increased N-cadherin expression by flow cytometry. Our data suggest that DWJM could serve as an ECM-based model to study AML stem cell-like cell behavior and chemotherapy sensitivity., (Copyright © 2018 Elsevier Ltd. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
33. Correlation between markers of bone metabolism and vitamin D levels in patients with monoclonal gammopathy of undetermined significance (MGUS).
- Author
-
Lipe B, Kambhampati S, Veldhuizen PV, Yacoub A, Aljitawi O, and Mikhael J
- Subjects
- Biomarkers analysis, Bone and Bones drug effects, Cholecalciferol deficiency, Dietary Supplements, Disease Progression, Humans, Monoclonal Gammopathy of Undetermined Significance drug therapy, Monoclonal Gammopathy of Undetermined Significance metabolism, Prospective Studies, Vitamin D blood, Vitamin D Deficiency drug therapy, Vitamin D Deficiency epidemiology, Bone and Bones metabolism, Cholecalciferol therapeutic use, Monoclonal Gammopathy of Undetermined Significance pathology
- Published
- 2017
- Full Text
- View/download PDF
34. Combined deficiency of factor V and factor VIII: management during cardiothoracic surgery.
- Author
-
Howard C and Lipe B
- Subjects
- Adult, Blood Coagulation Tests, Humans, Male, Factor V Deficiency drug therapy, Hemophilia A drug therapy, Thoracic Surgery methods
- Abstract
: Combined factor V and factor VIII deficiency is an extremely rare bleeding disorder for which research is lacking. We present the case of a 33-year-old man requiring repeat mitral valve replacement. A multidisciplinary team approach was utilized to minimize his risk of bleeding which included the use of plasma exchange, intravenous factor replacement, and platelet transfusion. This approach created an operative experience that did not require blood transfusion or the use of other hemostatic medications.
- Published
- 2017
- Full Text
- View/download PDF
35. Erythropoietin modulation is associated with improved homing and engraftment after umbilical cord blood transplantation.
- Author
-
Aljitawi OS, Paul S, Ganguly A, Lin TL, Ganguly S, Vielhauer G, Capitano ML, Cantilena A, Lipe B, Mahnken JD, Wise A, Berry A, Singh AK, Shune L, Lominska C, Abhyankar S, Allin D, Laughlin M, McGuirk JP, and Broxmeyer HE
- Subjects
- ADP-ribosyl Cyclase 1 metabolism, Adolescent, Adult, Aged, Animals, Antigens, CD34 metabolism, Cell Differentiation drug effects, Cell Movement drug effects, Chemokine CXCL12 pharmacology, Chimerism, Cohort Studies, Female, Graft vs Host Disease etiology, Humans, Hyperbaric Oxygenation, Male, Mice, Middle Aged, Myeloid Cells cytology, Myeloid Cells drug effects, Myeloid Cells metabolism, Receptors, Erythropoietin metabolism, Survival Analysis, Transplantation Conditioning, Treatment Outcome, Young Adult, Cord Blood Stem Cell Transplantation adverse effects, Erythropoietin metabolism
- Abstract
Umbilical cord blood (UCB) engraftment is in part limited by graft cell dose, generally one log less than that of bone marrow (BM)/peripheral blood (PB) cell grafts. Strategies toward increasing hematopoietic stem/progenitor cell (HSPC) homing to BM have been assessed to improve UCB engraftment. Despite recent progress, a complete understanding of how HSPC homing and engraftment are regulated is still elusive. We provide evidence that blocking erythropoietin (EPO)-EPO receptor (R) signaling promotes homing to BM and early engraftment of UCB CD34
+ cells. A significant population of UCB CD34+ HSPC expresses cell surface EPOR. Exposure of UCB CD34+ HSPC to EPO inhibits their migration and enhances erythroid differentiation. This migratory inhibitory effect was reversed by depleting EPOR expression on HSPC. Moreover, systemic reduction in EPO levels by hyperbaric oxygen (HBO) used in a preclinical mouse model and in a pilot clinical trial promoted homing of transplanted UCB CD34+ HSPC to BM. Such a systemic reduction of EPO in the host enhanced myeloid differentiation and improved BM homing of UCB CD34+ cells, an effect that was overcome with exogenous EPO administration. Of clinical relevance, HBO therapy before human UCB transplantation was well-tolerated and resulted in transient reduction in EPO with encouraging engraftment rates and kinetics. Our studies indicate that systemic reduction of EPO levels in the host or blocking EPO-EPOR signaling may be an effective strategy to improve BM homing and engraftment after allogeneic UCB transplantation. This clinical trial was registered at www.ClinicalTrials.gov (#NCT02099266)., (© 2016 by The American Society of Hematology.)- Published
- 2016
- Full Text
- View/download PDF
36. A pharmacokinetics and safety phase 1/1b study of oral ixazomib in patients with multiple myeloma and severe renal impairment or end-stage renal disease requiring haemodialysis.
- Author
-
Gupta N, Hanley MJ, Harvey RD, Badros A, Lipe B, Kukreti V, Berdeja J, Yang H, Hui AM, Qian M, Zhang X, Venkatakrishnan K, and Chari A
- Subjects
- Administration, Oral, Adult, Aged, Aged, 80 and over, Anemia etiology, Blood Proteins metabolism, Boron Compounds administration & dosage, Boron Compounds metabolism, Drug Dosage Calculations, Female, Glycine administration & dosage, Glycine metabolism, Glycine pharmacokinetics, Humans, Kidney Failure, Chronic therapy, Male, Middle Aged, Multiple Myeloma complications, Protease Inhibitors administration & dosage, Renal Dialysis, Boron Compounds pharmacokinetics, Glycine analogs & derivatives, Multiple Myeloma drug therapy, Protease Inhibitors pharmacokinetics, Renal Insufficiency therapy
- Abstract
Renal impairment (RI) is a major complication of multiple myeloma (MM). This study aimed to characterize the single-dose pharmacokinetics (PK) of the oral proteasome inhibitor, ixazomib, in cancer patients with normal renal function [creatinine clearance (CrCl) ≥90 ml/min; n = 20), severe RI (CrCl <30 ml/min; n = 14), or end-stage renal disease requiring haemodialysis (ESRD; n = 7). PK and adverse events (AEs) were assessed after a single 3 mg dose of ixazomib. Ixazomib was highly bound to plasma proteins (~99%) in all renal function groups. Unbound and total systemic exposures of ixazomib were 38% and 39% higher, respectively, in severe RI/ESRD patients versus patients with normal renal function. Total ixazomib concentrations were similar in pre- and post-dialyser samples collected from ESRD patients; therefore, ixazomib can be administered without regard to haemodialysis timing. Except for anaemia, the incidence of the most common AEs was generally similar across groups, but grade 3 and 4 AEs were more frequent in the severe RI/ESRD groups versus the normal group (79%/57% vs. 45%), as were serious AEs (43%/43% vs. 15%). The PK and safety results support a reduced ixazomib dose of 3 mg in patients with severe RI/ESRD., (© 2016 The Authors. British Journal of Haematology published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
37. Tracheal TDx fetal lung maturity test for assessing lung maturity in newborns with respiratory distress.
- Author
-
Giacoia GP, Lipe BH, Kelly C, and West K
- Subjects
- Fetal Organ Maturity, Humans, Infant, Newborn, Lung physiology, Pulmonary Surfactants analysis, Respiration, Artificial, Respiratory Distress Syndrome, Newborn therapy, Lung embryology, Respiratory Distress Syndrome, Newborn diagnosis, Sputum chemistry, Trachea chemistry
- Abstract
We evaluated a modified TDx-fetal lung maturity (FLM) test for estimating surfactant in small volume tracheal aspirates in 140 infants requiring mechanical ventilation. Respiratory distress syndrome was present in 75 infants, and 68 had TDx FLM less than 60 mg/dL (sensitivity, 90.6%). Respiratory distress was absent in 65 infants, 56 had TDx FLM more than 60 mg/dL (specificity, 86.1%). The modified TDx-FLM assay is highly automated, has a fast turn-around time (less than 30 minutes), has high reproducibility, and is less expensive than other available methods. This method has potential routine application in the diagnosis of surfactant deficiency, and the assessment of surfactant replacement and mechanical ventilation therapies.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.