1
|
Labanca C, Martino EA, Vigna E, Bruzzese A, Mendicino F, Lucia E, Olivito V, Puccio N, Neri A, Morabito F, Gentile M. Talquetamab in Multiple Myeloma: Efficacy, Safety, and Future Directions. Eur J Haematol 2025; 114:386-399. [PMID: 39604778 PMCID: PMC11798766 DOI: 10.1111/ejh.14353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
Relapsed and refractory multiple myeloma (RRMM) remains a challenging condition despite advances in immunotherapies. Novel bispecific antibodies (BsAbs), including talquetamab, have shown promising efficacy in heavily pretreated patients, even those with triple- and penta-refractory disease. Talquetamab, recently approved by the FDA and EMA, is indicated for patients who have progressed after at least three or four prior lines of therapy (LOTs). Administered following a step-up dosing phase to manage cytokine release syndrome (CRS), talquetamab demonstrated a high overall response rate (ORR) of approximately 70%, including in patients previously treated with T-cell redirecting therapies. Its safety profile is consistent with other BsAbs, with hematologic adverse events such as anemia and neutropenia commonly reported, alongside unique on-target off-tumor toxicities like dysgeusia and skin-related events. Infections were less frequent compared to other BsAbs. The optimal sequencing of talquetamab and other therapies, including CAR-T cell treatments, remains an area of active research, as resistance to anti-BCMA therapies presents ongoing clinical challenges. Current trials are exploring the use of talquetamab in combination therapies, as well as therapeutic strategies post-treating progression. The real-world data further support talquetamab's efficacy, making it a valuable addition to the RRMM treatment landscape.
Collapse
Affiliation(s)
| | | | - Ernesto Vigna
- Hematology UnitAzienda Ospedaliera AnnunziataCosenzaItaly
| | | | | | - Eugenio Lucia
- Hematology UnitAzienda Ospedaliera AnnunziataCosenzaItaly
| | | | - Noemi Puccio
- Laboratorio di Ricerca Traslazionale Azienda USL‐IRCSS Reggio EmiliaEmilia‐RomagnaItaly
| | - Antonino Neri
- Scientific Directorate IRCCS of Reggio EmiliaReggio EmiliaEmilia‐RomagnaItaly
| | | | - Massimo Gentile
- Hematology UnitAzienda Ospedaliera AnnunziataCosenzaItaly
- Department of Pharmacy, Health and Nutritional ScienceUniversity of CalabriaRendeItaly
| |
Collapse
|
2
|
Lloyd K, Middelburg J, Ovcinnikovs V, Pencheva N, Kemper K, van Hall T. Improving CD3 bispecific antibody therapy in solid tumors using combination strategies. Front Oncol 2025; 15:1548446. [PMID: 39995843 PMCID: PMC11847677 DOI: 10.3389/fonc.2025.1548446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/14/2025] [Indexed: 02/26/2025] Open
Abstract
CD3 bispecific antibodies (bsAbs) are emerging as an important treatment option in the arsenal of oncologists. There are numerous FDA-approved CD3 bsAbs for both hematological and solid tumors. Despite these recent advances, the success of CD3 bsAbs in solid cancer has been hampered by hurdles like limited intratumoral T cell numbers, immunosuppressive tumor microenvironments (TME), and poor memory T-cell induction. Furthermore, tumor surface antigen selection for an optimal therapeutic window and acceptable collateral damage to normal tissues is challenging. In this review, we discuss recent research investigating combination approaches aimed at improving CD3 bsAb efficacy in solid cancer.
Collapse
Affiliation(s)
| | - Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
3
|
Mouhieddine TH, Costa BA, Richter J. Advancements in bispecific antibodies for multiple myeloma: What's new and what lies ahead. Semin Hematol 2025; 62:58-70. [PMID: 39880754 DOI: 10.1053/j.seminhematol.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/04/2024] [Accepted: 12/22/2024] [Indexed: 01/31/2025]
Abstract
Recent advancements in multiple myeloma (MM) treatment-including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, and T cell-redirecting therapies like chimeric antigen receptor (CAR) T cells and bispecific antibodies (BsAbs)-have significantly improved patient outcomes. However, MM remains incurable, highlighting the need for novel therapeutic strategies. BsAbs, which simultaneously target a tumor-specific antigen and CD3 on T cells, have shown promising efficacy. Three BsAbs - teclistamab, elranatamab, and talquetamab - have been approved for relapsed or refractory MM, demonstrating response rates of 60 %-74 % and median progression-free survival of approximately 1 year. This review provides a comprehensive overview of the latest advancements in BsAb therapy for MM, focusing on new therapeutic targets such as BCMA, GPRC5D, and FcRH5, recent clinical trial data, safety considerations, and future directions. We discuss tumor-intrinsic mechanisms of resistance, including antigen expression variability and antigen escape, as well as immune-related factors like T-cell exhaustion and an immunosuppressive microenvironment. Future strategies involve integrating BsAbs earlier in treatment, combining them with other agents to enhance efficacy and overcome resistance, and optimizing administration protocols to mitigate adverse effects. By examining these developments, we highlight how BsAbs are reshaping the treatment landscape of MM and underscore the importance of ongoing research to improve survival and quality of life for patients.
Collapse
Affiliation(s)
- Tarek H Mouhieddine
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Bruno Almeida Costa
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joshua Richter
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
4
|
Middelburg J, Schaap G, Sluijter M, Lloyd K, Ovcinnikovs V, Schuurman J, van der Burg SH, Kemper K, van Hall T. Cancer vaccines compensate for the insufficient induction of protective tumor-specific immunity of CD3 bispecific antibody therapy. J Immunother Cancer 2025; 13:e010331. [PMID: 39800374 PMCID: PMC11749218 DOI: 10.1136/jitc-2024-010331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND CD3 bispecific antibody (CD3 bsAb) therapy has become an established treatment modality for some cancer types and exploits endogenous T cells irrespective of their specificity. However, durable clinical responses are hampered by immune escape through loss of tumor target antigen expression. Induction of long-lasting tumor-specific immunity might therefore improve therapeutic efficacy, but has not been studied in detail yet for CD3 bsAbs. Here, we examined multiple combination strategies aiming to improve survival rates in solid tumors and, simultaneously, install endogenous immunity capable of protection to tumor rechallenge. METHODS Two syngeneic mouse tumor models were employed: The immunologically "cold" B16F10 melanoma and the immunologically "hot" MC38.TRP1 colon carcinoma model. Mice were treated with CD3xTRP1 bsAb (murine Fc-inert immunoglobulin G2a) as monotherapy, or in combination with agonistic costimulatory antibodies, Fc-active tumor-opsonizing antibodies, or tumor-(non)specific vaccines. Treatment efficacy of primary tumors and protection from rechallenge was monitored, as well as induction of tumor-specific T-cell responses. RESULTS In the immunologically "cold" B16F10 model, all combination therapies improved antitumor activity compared with CD3 bsAb monotherapy and induced systemic tumor-specific T-cell responses. However, this endogenous T-cell immunity swiftly waned and failed to protect mice from subsequent tumor rechallenge, except for combination therapy with tumor-specific vaccination. These vaccines strongly improved the therapeutic efficacy of CD3 bsAb against primary tumors and led to long-term immunological protection. In the immunologically "hot" MC38.TRP1 model, CD3 bsAb combined with only the vaccine adjuvant was sufficient to generate protective T-cell immunity and, moreover, prevented tumor escape via antigen loss. CONCLUSIONS These results demonstrate the impact of tumor antigenicity on the induction of protective endogenous antitumor immunity during CD3 bsAb treatment and, importantly, show that the combination with tumor-specific vaccines improves therapeutic efficacy and installs long-term immunological memory in both "hot" and "cold" tumors.
Collapse
Affiliation(s)
- Jim Middelburg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Gaby Schaap
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Marjolein Sluijter
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Katy Lloyd
- Genmab BV, Utrecht, Utrecht, The Netherlands
| | | | | | - Sjoerd H van der Burg
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Thorbald van Hall
- Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
5
|
Lee H, Durante M, Skerget S, Vishwamitra D, Benaoudia S, Ahn S, Poorebrahim M, Barakat E, Jung D, Leblay N, Ziccheddu B, Diamond B, Papadimitriou M, Cohen AD, Landgren O, Neri P, Maura F, Bahlis NJ. Impact of soluble BCMA and non-T-cell factors on refractoriness to BCMA-targeting T-cell engagers in multiple myeloma. Blood 2024; 144:2637-2651. [PMID: 39321344 PMCID: PMC11738017 DOI: 10.1182/blood.2024026212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
ABSTRACT Adoptive T-cell therapy is a promising therapy for multiple myeloma (MM), but its efficacy hinges on understanding the relevant biologic and predictive markers of response. B-cell maturation antigen (BCMA) is a key target antigen in MM with active development of multiple anti-BCMA T-cell engagers (TCEs) and chimeric antigen receptor T-cell therapies. The regulation of surface BCMA expression by MM cells, which leads to shedding of soluble BCMA (sBCMA), has triggered debate about the significance of sBCMA as a predictive marker and its potential impact on treatment outcomes. To address this, we leveraged whole-genome sequencing and in vitro assays to demonstrate that sBCMA may independently predict primary refractoriness to anti-BCMA therapies. In addition to sBCMA, tumor burden and surface BCMA antigen density collectively influenced the anti-BCMA TCE cytotoxic efficacy. Correlative analyses of 163 patients treated with the anti-BCMA TCE teclistamab validated and further underscored the association between elevated baseline sBCMA (>400 ng/mL) and refractoriness. Importantly, increasing the TCE dose, using TCE against alternative targets (eg, GPRC5D), and gamma secretase inhibitors were able to overcome the high sBCMA levels. These findings highlight the importance of taking into account the baseline sBCMA levels, disease burden, and TCE dose intensity when administering anti-BCMA TCEs, thereby offering critical insights for optimizing therapeutic strategies to overcome specific high-risk features and primary anti-BCMA TCE refractoriness.
Collapse
Affiliation(s)
- Holly Lee
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Michael Durante
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | | | | | - Sacha Benaoudia
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Sungwoo Ahn
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Mansour Poorebrahim
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Elie Barakat
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - David Jung
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Noémie Leblay
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Bachisio Ziccheddu
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | - Benjamin Diamond
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | - Marios Papadimitriou
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | - Adam D. Cohen
- Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Ola Landgren
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Francesco Maura
- Sylvester Comprehensive Cancer Center, Myeloma Research Institute, University of Miami, Miami, FL
| | - Nizar J. Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
6
|
Zweegman S. Supportive care in myeloma-when treating the clone alone is not enough. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:569-581. [PMID: 39643986 PMCID: PMC11665544 DOI: 10.1182/hematology.2024000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
The overall survival in patients with multiple myeloma has increased over recent decades. This trend is anticipated to further advance with the emergence of T-cell-redirecting therapies, including chimeric antigen receptor T-cell (CAR T) therapy and T-cell-engaging bispecific antibodies. Despite these therapeutic improvements, treatment-related adverse events impede quality of life. This underscores the imperative of optimizing supportive care strategies to maximize treatment outcomes. Such optimization is crucial not only for patient well-being but also for treatment adherence, which may translate into long-term disease control. We here describe a) how to prevent bone disease, b) a risk-adapted thrombosis prophylaxis approach, c) the management of on-target, off-tumor toxicity of G-protein-coupled receptor class C group 5 member D-targeting T-cell-redirecting therapies, and d) infectious prophylaxis, with a focus on infections during T-cell-redirecting therapies.
Collapse
Affiliation(s)
- Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Center, Universiteit van Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Jiang H, Li L, Guo M, Li M, Wu H, Chen X, Gao M, Xu Q, Mi J, Cui C, Fu W. Efficacy of daratumumab on multiple myeloma patients with renal insufficiency: a systematic review and meta-analysis. Hematology 2024; 29:2399430. [PMID: 39248713 DOI: 10.1080/16078454.2024.2399430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND Renal insufficiency (RI) is a key factor affecting the prognosis of multiple myeloma (MM) patients. Because the benefit of daratumumab for treating MM patients with RI remains unclear, our objective was to evaluate the efficacy of daratumumab on MM patients with RI. METHODS We conducted a systematic search of the PubMed, EMBASE, and Cochrane Library databases as of October 24, 2023. Two independent reviewers screened the article titles, abstracts, and full text to identify the randomized controlled trials (RCTs) meeting the inclusion and exclusion criteria. Meta-analyses were performed using RevMan version 5.4. Outcomes of interest were progression-free survival (PFS), overall survival (OS), complete response or better (≥CR), and minimal residual disease (MRD) negativity, all calculated as hazard ratios (HRs) or risk ratios (RRs) with 95% confidence intervals (CIs). RESULTS A total of 10 RCTs with 5003 patients were included. Add-on daratumumab improved PFS and OS among newly diagnosed MM (NDMM) patients with RI (HR 0.48 [95% CI: 0.36, 0.64, I2 = 65%] and HR 0.63 [95% CI: 0.48, 0.82, I2 = 0%]) as well as relapsed/refractory MM (RRMM)-RI patients, compared with the control group (HR 0.46 [95% CI: 0.37, 0.58, I2 = 0%] and HR 0.68 [95% CI: 0.51, 0.92, I2 = 0%]). In terms of the renal status, the efficacy of add-on daratumumab for MMRI patients was similar to that for MM patients with normal renal function. A prolonged PFS benefit for add-on daratumumab treatment versus the control was evident across all RRMM-RI subgroups, and the benefits tended to increase with the follow-up time. CONCLUSIONS Our results indicate that MM patients with RI could benefit from a daratumumab-added regimen regardless of MM status. Additional high-quality RCTs are still warranted to confirm our findings.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Lu Li
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Meiyu Guo
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Meizhang Li
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Hao Wu
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Xiaomei Chen
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Mingzhao Gao
- Medical Affairs, Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Qianqian Xu
- Medical Affairs, Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Jia Mi
- Medical Affairs, Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Canchan Cui
- Medical Affairs, Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Weijun Fu
- Department of Hematology, School of Medicine, Shanghai Fourth People's Hospital, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
8
|
van de Donk NW, Rasche L, Sidana S, Zweegman S, Garfall AL. T Cell-Redirecting Bispecific Antibodies in Multiple Myeloma: Optimal Dosing Schedule and Duration of Treatment. Blood Cancer Discov 2024; 5:388-399. [PMID: 39321136 PMCID: PMC11528190 DOI: 10.1158/2643-3230.bcd-24-0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/22/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024] Open
Abstract
T cell-redirecting bispecific antibodies (BsAb) induce significant responses in heavily pretreated multiple myeloma. BsAbs are currently administered in a dose-dense manner until disease progression. However, continuous therapy is associated with safety concerns, including a high risk of infections and high costs. In addition, chronic exposure to BsAbs, and thus long-term T-cell stimulation, induces T-cell exhaustion, which may contribute to relapse. There is increasing evidence that the strategy of induction treatment followed by maintenance with longer intervals between BsAb doses, or limited treatment duration with cessation of therapy in patients who achieve deep remission, improves the balance between toxicity and efficacy. Significance: There is increasing evidence that after initial debulking, less-frequent BsAb administration mitigates T-cell exhaustion and minimizes the potential for chronic or cumulative toxicity while maintaining durable clinical responses. In addition, specific patient subsets may experience an extended treatment-free period following fixed-duration treatment. Fixed-duration treatment may, therefore, decrease cumulative toxicities and the burden on patients and healthcare systems.
Collapse
Affiliation(s)
- Niels W.C.J. van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Leo Rasche
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, California
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| | - Alfred L. Garfall
- Division of Hematology and Oncology, Department of Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
9
|
Rees M, Abdallah N, Yohannan B, Gonsalves WI. Bispecific antibody targets and therapies in multiple myeloma. Front Immunol 2024; 15:1424925. [PMID: 39450163 PMCID: PMC11499143 DOI: 10.3389/fimmu.2024.1424925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Recently, several bispecific antibodies (BsAbs) have been approved for the treatment of relapsed multiple myeloma (MM) after early phase trials in heavily pre-treated patients demonstrated high response rates and impressive progression-free survival with monotherapy. These BsAbs provide crucial treatment options for relapsed patients and challenging decisions for clinicians. Evidence on the optimal patient population, treatment sequence, and duration of these therapeutics is unknown and subject to active investigation. While rates of cytokine release syndrome and neurotoxicity appear to be lower with BsAbs than with CAR T-cells, morbidity from infection is high and novel pathways of treatment resistance arise from the longitudinal selection pressure of chronic BsAb therapy. Lastly, a wealth of novel T-cell engagers with unique antibody-structures and antigenic targets are under active investigation with promising early outcome data. In this review, we examine the mechanism of action, therapeutic targets, combinational approaches, sequencing and mechanisms of disease relapse for BsAbs in MM.
Collapse
Affiliation(s)
- Matthew Rees
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, St Vincent’s Hospital Melbourne,
Melbourne, VIC, Australia
| | - Nadine Abdallah
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
| | - Binoy Yohannan
- Division of Hematology, Mayo Clinic, Rochester, MN, United States
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | | |
Collapse
|
10
|
Devasia AJ, Chari A, Lancman G. Bispecific antibodies in the treatment of multiple myeloma. Blood Cancer J 2024; 14:158. [PMID: 39266530 PMCID: PMC11393350 DOI: 10.1038/s41408-024-01139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
The treatment paradigm in myeloma is constantly changing. Upfront use of monoclonal antibodies like daratumumab along with proteasome inhibitors (PI)s, and immune modulators (IMiD)s have significantly improved survival and outcomes, but also cause unique challenges at the time of relapse. Engaging immune T cells for tumour cell kill with chimeric antigenic T-cell (CAR T-cell) therapy and bispecific antibodies have become important therapeutic options in relapsed multiple myeloma. Bispecific antibodies are dual antigen targeting constructs that engage the T cells to plasma cells through various target antigens like B-cell membrane antigen (BCMA), G-protein-coupled receptor family C group 5 member D (GPRC5D), and Fc receptor-homolog 5 (FcRH5). These agents have proven to induce deep and durable responses in heavily pre-treated myeloma patients with a predictable safety profile and the ease of off-the-shelf availability. Significant research is ongoing to overcome resistance mechanisms like T cell exhaustion, target antigen mutation or loss and high disease burden. Various trials are also studying these agents as first line options in the newly diagnosed setting. These agents play an important role in the relapsed setting, and efforts are underway to optimize their sequencing in the myeloma treatment algorithm.
Collapse
Affiliation(s)
| | - Ajai Chari
- University of California, San Francisco, San Francisco, CA, USA
| | - Guido Lancman
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
11
|
van de Donk NWCJ, Chari A, Mateos MV. Mechanisms of resistance against T-cell engaging bispecific antibodies in multiple myeloma: implications for novel treatment strategies. Lancet Haematol 2024; 11:e693-e707. [PMID: 39033769 DOI: 10.1016/s2352-3026(24)00186-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 07/23/2024]
Abstract
Off-the-shelf T-cell-redirecting bispecific antibodies targeting BCMA, GPRC5D, and FcRH5 have high activity in multiple myeloma with a manageable toxicity profile. However, not all patients respond to bispecific antibodies and patients can develop bispecific antibody resistance after an initial response. Mechanisms that contribute to bispecific antibody resistance are multifactorial and include tumour-related factors, such as high tumour burden, expression of T-cell inhibitory ligands, and antigen loss. Resistance due to antigen escape can be prevented by simultaneously targeting two tumour-associated antigens with a trispecific antibody or a combination of two bispecific antibodies. There is also increasing evidence that primary resistance to bispecific antibodies is associated with impaired baseline T-cell function. Long-term exposure to bispecific antibodies with chronic T-cell stimulation further aggravates T-cell dysfunction, which could contribute to failure of disease control. Therapeutic interference with T-cell exhaustion by targeting inhibitory or costimulatory pathways could improve bispecific antibody-mediated antitumour activity. The immunosuppressive microenvironment also contributes to bispecific antibody resistance. CD38-targeting antibodies hold promise as combination partners for bispecific antibodies because of their potential to eliminate CD38+ immune suppressor cells. In conclusion, a better understanding of the mechanisms underlying the absence of disease response has provided novel insights to optimise T-cell activity and bispecific antibody efficacy in multiple myeloma.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, Netherlands.
| | - Ajai Chari
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Maria Victoria Mateos
- Instituto de Investigación Biomédica de Salamanca, Centro de Investigación del Cáncer, Centro de Investigación Biomédica en Red Cáncer, University Hospital of Salamanca, Salamanca, Spain
| |
Collapse
|
12
|
Heerma van Voss MR, Molenaar RJ, Korst CLBM, Bartelink IH, Baglio SR, Kruyswijk S, de Ruijter M, Zweegman S, Kuipers MT, van de Donk NWCJ. T-cell redirecting bispecific antibody treatment in multiple myeloma: current knowledge and future strategies for sustained T-cell engagement. Expert Opin Biol Ther 2024; 24:889-901. [PMID: 39185748 DOI: 10.1080/14712598.2024.2397436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
INTRODUCTION T-cell redirecting bispecific antibodies (BsAbs), targeting B-cell maturation antigen (BCMA) or G-protein - coupled receptor class C group 5 member D (GPRC5D), are efficacious new agents for the treatment of patients with relapsed or refractory MM. AREAS COVERED This review discusses the pharmacokinetic properties, efficacy, and safety profile of T-cell redirecting BsAbs in MM, with a special focus on their optimal dosing schedule, resistance mechanisms and future strategies to enhance efficacy, reduce toxicity, and maximize duration of response. EXPERT OPINION To further improve the efficacy of BsAbs, ongoing studies are investigating whether combination therapy can enhance depth and duration of response. An important open question is also to what extent response to BsAbs can be improved when these agents are used in earlier lines of therapy. In addition, more evidence is needed on rational de-intensification strategies of BsAb dosing upon achieving a sufficient response, and if (temporary) treatment cessation is possible in patients who have achieved a deep remission (e.g. complete response or minimal residual disease-negative status).
Collapse
Affiliation(s)
- Marise R Heerma van Voss
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Remco J Molenaar
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Charlotte L B M Korst
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Imke H Bartelink
- Department of Pharmacy and Clinical Pharmacology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Serena R Baglio
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maaike de Ruijter
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maria T Kuipers
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Verkleij CPM, O'Neill CA, Broekmans MEC, Frerichs KA, Bruins WSC, Duetz C, Kruyswijk S, Baglio SR, Skerget S, Montes de Oca R, Zweegman S, Verona RI, Mutis T, van de Donk NWCJ. T-Cell Characteristics Impact Response and Resistance to T-Cell-Redirecting Bispecific Antibodies in Multiple Myeloma. Clin Cancer Res 2024; 30:3006-3022. [PMID: 38687588 DOI: 10.1158/1078-0432.ccr-23-3333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/15/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE Bispecific antibodies (BsAb) directed against B-cell maturation antigen (teclistamab) or the orphan G protein-coupled receptor GPRC5D (talquetamab) induce deep and durable responses in heavily pretreated patients with multiple myeloma. However, mechanisms underlying primary and acquired resistance remain poorly understood. EXPERIMENTAL DESIGN The anti-multiple myeloma activity of teclistamab and talquetamab was evaluated in bone marrow (BM) samples from patients with multiple myeloma. T-cell phenotype and function were assessed in BM/peripheral blood samples obtained from patients with multiple myeloma who were treated with these BsAb. RESULTS In ex vivo killing assays with 41 BM samples from BsAb-naive patients with multiple myeloma, teclistamab- and talquetamab-mediated multiple myeloma lysis was strongly correlated (r = 0.73, P < 0.0001). Both BsAb exhibited poor activity in samples with high regulatory T-cell (Treg) numbers and a low T-cell/multiple myeloma cell ratio. Furthermore, comprehensive phenotyping of BM samples derived from patients treated with teclistamab or talquetamab revealed that high frequencies of PD-1+ CD4+ T cells, CTLA4+ CD4+ T cells, and CD38+ CD4+ T cells were associated with primary resistance. Although this lack of response was linked to a modest increase in the expression of inhibitory receptors, increasing T-cell/multiple myeloma cell ratios by adding extra T cells enhanced sensitivity to BsAb. Further, treatment with BsAb resulted in an increased proportion of T cells expressing exhaustion markers (PD-1, TIGIT, and TIM-3), which was accompanied by reduced T-cell proliferative potential and cytokine secretion, as well as impaired antitumor efficacy in ex vivo experiments. CONCLUSIONS Primary resistance is characterized by a low T-cell/multiple myeloma cell ratio and Treg-driven immunosuppression, whereas reduced T-cell fitness due to continuous BsAb-mediated T-cell activation may contribute to the development of acquired resistance.
Collapse
Affiliation(s)
- Christie P M Verkleij
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Chloe A O'Neill
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Marloes E C Broekmans
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Kristine A Frerichs
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Wassilis S C Bruins
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Carolien Duetz
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Serena R Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sheri Skerget
- Janssen Research & Development, Spring House, Pennsylvania
| | | | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
14
|
Żyłka K, Kubicki T, Gil L, Dytfeld D. T-cell exhaustion in multiple myeloma. Expert Rev Hematol 2024; 17:295-312. [PMID: 38919090 DOI: 10.1080/17474086.2024.2370552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Chimeric Antigen Receptor (CAR) T-cells and Bispecific Antibodies (BsAb) are the leading platforms for redirecting the immune system against cells expressing the specific antigen, revolutionizing the treatment of hematological malignancies, including multiple myeloma (MM). In MM, drug-resistant relapses are the main therapy-limiting factor and the leading cause of why the disease is still considered incurable. T-cell-engaging therapies hold promise in improving the treatment of MM. However, the effectiveness of these treatments may be hindered by T-cell fitness. T-cell exhaustion is a condition of a gradual decline in effector function, reduced cytokine secretion, and increased expression of inhibitory receptors due to chronic antigen stimulation. AREAS COVERED This review examines findings about T-cell exhaustion in MM in the context of T-cell redirecting BsAbs and CAR-T treatment. EXPERT OPINION The fitness of T-cells has become an important factor in the development of T-cell redirecting therapies. The way T-cell exhaustion relates to these therapies could affect the further development of CAR and BsAbs technologies, as well as the strategies used for clinical use. Therefore, this review aims to explore the current understanding of T-cell exhaustion in MM and its relationship to these therapies.
Collapse
Affiliation(s)
- Krzysztof Żyłka
- The Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznań, Poland
| | - Tadeusz Kubicki
- The Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznań, Poland
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Lidia Gil
- The Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznań, Poland
| | - Dominik Dytfeld
- The Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
15
|
Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A, Zhou S. Cancer therapy with antibodies. Nat Rev Cancer 2024; 24:399-426. [PMID: 38740967 PMCID: PMC11180426 DOI: 10.1038/s41568-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Maximilian F Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Katharine M Wright
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA
| | - Sandra B Gabelli
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA.
| | - Mitchell Ho
- Antibody Engineering Program, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Rodriguez-Otero P, Usmani S, Cohen AD, van de Donk NWCJ, Leleu X, Gállego Pérez-Larraya J, Manier S, Nooka AK, Mateos MV, Einsele H, Minnema M, Cavo M, Derman BA, Puig N, Gay F, Ho PJ, Chng WJ, Kastritis E, Gahrton G, Weisel K, Nagarajan C, Schjesvold F, Mikhael J, Costa L, Raje NS, Zamagni E, Hájek R, Weinhold N, Yong K, Ye JC, Sidhana S, Merlini G, Martin T, Lin Y, Chari A, Popat R, Kaufman JL. International Myeloma Working Group immunotherapy committee consensus guidelines and recommendations for optimal use of T-cell-engaging bispecific antibodies in multiple myeloma. Lancet Oncol 2024; 25:e205-e216. [PMID: 38697166 DOI: 10.1016/s1470-2045(24)00043-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 05/04/2024]
Abstract
Multiple myeloma remains an incurable disease, despite the development of numerous drug classes and combinations that have contributed to improved overall survival. Immunotherapies directed against cancer cell-surface antigens, such as chimeric antigen receptor (CAR) T-cell therapy and T-cell-redirecting bispecific antibodies, have recently received regulatory approvals and shown unprecedented efficacy. However, these immunotherapies have unique mechanisms of action and toxicities that are different to previous treatments for myeloma, so experiences from clinical trials and early access programmes are essential for providing specific recommendations for management of patients, especially as these agents become available across many parts of the world. Here, we provide expert consensus clinical practice guidelines for the use of bispecific antibodies for the treatment of myeloma. The International Myeloma Working Group is also involved in the collection of prospective real-time data of patients treated with such immunotherapies, with the aim of learning continuously and adapting clinical practices to optimise the management of patients receiving immunotherapies.
Collapse
Affiliation(s)
| | - Saad Usmani
- Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Adam D Cohen
- University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
| | | | - Xavier Leleu
- Centre Hospitalier Universitaire Poitiers, Poitiers, France
| | - Jaime Gállego Pérez-Larraya
- Department of Neurology, Clínica Universidad de Navarra, Cancer Center Clinica Universidad de Navarra, CCUN, Pamplona, Spain
| | - Salomon Manier
- Centre Hospitalier Universitaire Lille, Universite de Lille, Lille, France
| | - Ajay K Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Maria Victoria Mateos
- University Hospital of Salamanca/IBSAL/Cancer Research Center-IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Hermann Einsele
- Universitätsklinikum Würzburg, Department of Internal Medicine II, Würzburg, Germany
| | - Monique Minnema
- Department of Hematology, University Medical Center, Utrecht, Netherlands
| | - Michele Cavo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Dipartimento di Scieze Mediche e Chirurgiche, Universitá di Bologna, Bologna, Italy
| | - Benjamin A Derman
- Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Noemi Puig
- University Hospital of Salamanca/IBSAL/Cancer Research Center-IBMCC (USAL-CSIC), CIBERONC, Salamanca, Spain
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, Torino, Italy
| | - P Joy Ho
- Royal Prince Alfred Hospital and University of Sydney, Sydney, NSW, Australia
| | - Wee-Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore
| | - Efstathios Kastritis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Gösta Gahrton
- Department of Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Katja Weisel
- Universitätsklinikum Hamburg-Eppendorf, Medizinische Klinik und Poliklinik, Hamburg, Germany
| | - Chandramouli Nagarajan
- Department of Haematology, Singapore General Hospital and SingHealth Duke NUS Blood Cancer Center, Singapore
| | - Fredik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital, Oslo, Norway; KG Jebsen Center for B Cell Malignancies, University of Oslo, Oslo, Norway
| | - Joseph Mikhael
- Translational Genomics Research Institute, City of Hope Cancer Center, Phoenix, AZ, USA; International Myeloma Foundation, Studio City, CA, USA
| | - Luciano Costa
- Department of Medicine, Division of Hematology and Oncology, University of Wisconsin, Madison, WI, USA
| | - Noopur S Raje
- Department of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy; Dipartimento di Scieze Mediche e Chirurgiche, Universitá di Bologna, Bologna, Italy
| | - Roman Hájek
- Department of Hemato-oncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Niels Weinhold
- Department of Medicine V, Multiple Myeloma Section, University Hospital Heidelberg, Heidelberg, Germany
| | - Kwee Yong
- University College London Hospitals, London, UK
| | | | - Surbhi Sidhana
- Stanford University School of Medicine, Stanford, CA, USA
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Tom Martin
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Yi Lin
- Mayo Clinic, Rochester, MN, USA
| | - Ajai Chari
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Rakesh Popat
- NIHR UCLH Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
17
|
Parrondo RD, Ailawadhi S, Cerchione C. Bispecific antibodies for the treatment of relapsed/refractory multiple myeloma: updates and future perspectives. Front Oncol 2024; 14:1394048. [PMID: 38660139 PMCID: PMC11039948 DOI: 10.3389/fonc.2024.1394048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Patients with relapsed/refractory multiple myeloma (RRMM) that are refractory to the five most active anti-MM drugs, so-called penta-refractory MM, have historically had dismal outcomes with subsequent therapies. Progressive immune dysfunction, particularly of the T-cell repertoire, is implicated in the development of disease progression and refractory disease. However, the advent of novel immunotherapies such as bispecific antibodies are rapidly changing the treatment landscape and improving the survival outcomes of patients with RRMM. Bispecific antibodies are antibodies that are engineered to simultaneously engage cytotoxic immune effector cells (T cells or NK cells) and malignant plasma cells via binding to immune effector cell antigens and extracellular plasma cell antigens leading to immune effector cell activation and malignant plasma cell destruction. Currently, bispecific antibodies that bind CD3 on T cells and plasma cell epitopes such as B-cell maturation antigen (BCMA), G-protein coupled receptor family C group 5 member D (GPRC5d), and Fc receptor homologue 5 (FcRH5) are the most advanced in clinical development and are showing unprecedented response rates in patients with RRMM, including patients with penta-refractory disease. In this review article, we explore the available clinical data of bispecific antibodies in RRMM and summarize the efficacy, safety, toxicity, clinical outcomes, mechanisms of resistance, and future directions of these therapies in patients with RRMM.
Collapse
Affiliation(s)
- Ricardo D. Parrondo
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Sikander Ailawadhi
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, and Cellular Therapies, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Claudio Cerchione
- Hematology Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
18
|
van de Donk NWCJ, Zweegman S. Monoclonal Antibodies in the Treatment of Multiple Myeloma. Hematol Oncol Clin North Am 2024; 38:337-360. [PMID: 38151402 DOI: 10.1016/j.hoc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
The incorporation of monoclonal antibodies into backbone regimens has substantially improved the clinical outcomes of patients with newly diagnosed and relapsed/refractory multiple myeloma (MM). Although the SLAMF7-targeting antibody elotuzumab has no single- agent activity, there is clinical synergy between elotuzumab and immunomodulatory drugs in patients with relapsed/refractory disease. Daratumumab and isatuximab are CD38-targeting antibodies which have single-agent activity and a favorable safety profile, which make these agents an attractive component of combination regimens. Monoclonal antibodies may cause infusion-related reactions, but with subcutaneous administration these are less frequently observed. All therapeutic antibodies may interfere with assessment of complete response. Next-generation Fc-engineered monoclonal antibodies are in development with the potential to further improve the outcome of patients with MM.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands.
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Lee H, Neri P, Bahlis NJ. BCMA- or GPRC5D-targeting bispecific antibodies in multiple myeloma: efficacy, safety, and resistance mechanisms. Blood 2024; 143:1211-1217. [PMID: 38194680 DOI: 10.1182/blood.2023022499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024] Open
Abstract
ABSTRACT Bispecific antibodies that engage T cells to target B-cell maturation antigen or G-protein-coupled receptor class C group 5 member D have demonstrated remarkable efficacy in heavily pretreated relapsed or refractory multiple myeloma (MM), leading to the recent accelerated approval of teclistamab, elranatamab, and talquetamab by health agencies. Future challenges, however, remain to define their optimal dosing schedule and duration, sequencing, and integration with established anti-MM therapeutics as well as delineating the biological and clinical mediators of immune escape.
Collapse
Affiliation(s)
- Holly Lee
- Department of Medicine, Divisions of Hematology and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Paola Neri
- Department of Medicine, Divisions of Hematology and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Nizar J Bahlis
- Department of Medicine, Divisions of Hematology and Oncology, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Tomita U, Ishimoto Y, Ri M, Kawase Y, Hizukuri Y, Maru C, Nanai K, Nakamura R, Nakayama M, Oguchi-Oshima K, Sumi H, Ohtsuka T, Iida S, Agatsuma T. A novel T cell-redirecting anti-GPRC5D × CD3 bispecific antibody with potent antitumor activity in multiple myeloma preclinical models. Sci Rep 2024; 14:5135. [PMID: 38429446 PMCID: PMC10907593 DOI: 10.1038/s41598-024-55143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
G-protein-coupled receptor class 5 member D (GPRC5D) is detected in malignant plasma cells in approximately 90% of patients diagnosed with multiple myeloma (MM). Here, we constructed BsAb5003, a novel humanized bispecific monoclonal antibody targeting CD3 and GPRC5D, and evaluated its therapeutic impact on MM. BsAb5003 induced specific cytotoxicity of GPRC5D-positive MM cells with concomitant T cell activation and cytokine release. The efficacy of BsAb5003 was associated with GPRC5D expression levels in MM cell lines. Flow cytometry analysis of bone marrow mononuclear cells (BMMNCs) from 49 MM patients revealed that GPRC5D was expressed in a wide population of MM patients, including heavily treated and high-risk patients. In ex vivo assays using BMMNCs, BsAb5003 induced potent efficacy against CD138 + MM cells in both newly diagnosed and relapsed/refractory patient samples in a GPRC5D expression-dependent manner. BsAb5003 significantly enhanced T cell activation and cytokine production in combination with immunomodulatory drugs (IMiDs) against MM cell lines. BsAb5003 also demonstrated significant inhibition of in vivo tumor growth by recruiting T cells. Taken together, these results suggest that T cell-redirecting bispecific antibody targeting GPRC5D as monotherapy and combination therapy with IMiDs could be a highly potent and effective treatment approach for a wide population of MM patients.
Collapse
Affiliation(s)
| | | | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | | | | | | | | | | | | | | | | | | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | |
Collapse
|
21
|
Martino EA, Bruzzese A, Labanca C, Mendicino F, Lucia E, Olivito V, Neri A, Morabito F, Vigna E, Gentile M. Teclistamab-cqyv in multiple myeloma. Eur J Haematol 2024; 112:320-327. [PMID: 37848191 DOI: 10.1111/ejh.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023]
Abstract
Multiple myeloma (MM) is an incurable neoplasm characterized by significant morbidity and mortality. Despite advances in treatment, MM patients eventually experienced a relapse of the disease. Penta-drug refractory patients continue to be the hard core of relapsed/refractory (RR) settings. Teclistamab-cqyv is a humanized IgG4 antibody and a bispecific BCMA-director CD3 T-cell engager. It recruits endogenous T cells, by targeting CD3 receptors expressed on their surface, resulting in their activation against BCMA, an antigen expressed by plasma cells. US Food and Drug Administration (FDA) and European Medicines Agency (EMA) have approved Teclistamab-cqyv in monotherapy for the treatment of RRMM patients who have received at least three prior therapies, including immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and anti-CD38 monoclonal antibodies (MoAbs) and have demonstrated disease progression during the last therapy. Its effectiveness was demonstrated in a pivotal clinical trial where the overall response rate (ORR) reached 60%. Other clinical studies are currently ongoing to investigate the association of the bispecific antibody with novel drugs with encouraging preliminary results, especially in the setting of heavily pretreated patients. In this review, the authors will provide a comprehensive overview of the drug, including its mechanism of action, major clinical trials, and future perspectives.
Collapse
Affiliation(s)
| | | | | | | | - Eugenio Lucia
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | | | - Antonino Neri
- Scientific Directorate IRCCS of Reggio Emilia, Reggio Emilia, Emilia-Romagna, Italy
| | | | - Ernesto Vigna
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
| | - Massimo Gentile
- Hematology Unit, Azienda Ospedaliera Annunziata, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
22
|
Frerichs KA, Verkleij CPM, Mateos MV, Martin TG, Rodriguez C, Nooka A, Banerjee A, Chastain K, Perales-Puchalt A, Stephenson T, Uhlar C, Kobos R, van der Holt B, Kruyswijk S, Kuipers MT, Groen K, Vishwamitra D, Skerget S, Cortes-Selva D, Doyle M, Zaaijer HL, Zweegman S, Verona RI, van de Donk NWCJ. Teclistamab impairs humoral immunity in patients with heavily pretreated myeloma: importance of immunoglobulin supplementation. Blood Adv 2024; 8:194-206. [PMID: 38052042 PMCID: PMC10787247 DOI: 10.1182/bloodadvances.2023011658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT Teclistamab and other B-cell maturation antigen (BCMA)-targeting bispecific antibodies (BsAbs) have substantial activity in patients with heavily pretreated multiple myeloma (MM) but are associated with a high rate of infections. BCMA is also expressed on normal plasma cells and mature B cells, which are essential for the generation of a humoral immune response. The aim of this study was to improve the understanding of the impact of BCMA-targeting BsAbs on humoral immunity. The impact of teclistamab on polyclonal immunoglobulins and B cell counts was evaluated in patients with MM who received once-weekly teclistamab 1.5 mg/kg subcutaneously. Vaccination responses were assessed in a subset of patients. Teclistamabinduced rapid depletion of peripheral blood B cells in patients with MM and eliminated normal plasma cells in ex vivo assays. In addition, teclistamab reduced the levels of polyclonal immunoglobulins (immunoglobulin G [IgG], IgA, IgE, and IgM), without recovery over time while receiving teclistamab therapy. Furthermore, response to vaccines against Streptococcus pneumoniae, Haemophilus influenzae type B, and severe acute respiratory syndrome coronavirus 2 was severely impaired in patients treated with teclistamab compared with vaccination responses observed in patients with newly diagnosed MM or relapsed/refractory MM. Intravenous immunoglobulin (IVIG) use was associated with a significantly lower risk of serious infections among patients treated with teclistamab (cumulative incidence of infections at 6 months: 5.3% with IVIG vs 54.8% with observation only [P < .001]). In conclusion, our data show severe defects in humoral immunity induced by teclistamab, the impact of which can be mitigated by the use of immunoglobulin supplementation. This trial was registered at www.ClinicalTrials.gov as #NCT04557098.
Collapse
Affiliation(s)
- Kristine A Frerichs
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | | | | | - Ajay Nooka
- Winship Cancer Institute, Emory University, Atlanta, GA
| | | | | | | | | | | | - Rachel Kobos
- Janssen Research & Development, Spring House, PA
| | - Bronno van der Holt
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Sandy Kruyswijk
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maria T Kuipers
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Kaz Groen
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | | | | | | | - Hans L Zaaijer
- Department of Medical Microbiology, Amsterdam UMC location, Academic Medical Center, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | | | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Lee H, Neri P, Bahlis NJ. Current use of bispecific antibodies to treat multiple myeloma. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2023; 2023:332-339. [PMID: 38066842 PMCID: PMC10727080 DOI: 10.1182/hematology.2023000433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Targeted immunotherapy has significantly improved the outcome of patients with hematological malignancies by leveraging the power of the immune system to eliminate tumor cells. In multiple myeloma (MM), bispecific T-cell engagers (BsAb) targeting B-cell maturation antigen (BCMA), G protein-coupled receptor, class C, group 5, member D (GPRC5D), and Fc receptor-like 5 (FcRL5) have already demonstrated remarkable clinical activity in triple-class refractory patients. However, responses to BsAb are not universal, and resistance often emerges while on therapy. Mechanisms mediating resistance are tumor intrinsic or immune dependent. Reported tumor intrinsic factors include antigenic loss (biallelic or functional) through deletions or mutations of target genes, increased soluble BCMA (for BCMA targeting BsAb), high tumor burden, and extramedullary disease. Immune-mediated resistance are largely dependent on T-cell fitness and tolerant immune environment. Understanding these mechanisms will allow the design of optimized BsAb therapy and an informed approach to sequencing and combining these molecules with other anti-MM agents and immune therapies.
Collapse
Affiliation(s)
- Holly Lee
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
24
|
Morè S, Corvatta L, Manieri VM, Morsia E, Poloni A, Offidani M. Novel Immunotherapies and Combinations: The Future Landscape of Multiple Myeloma Treatment. Pharmaceuticals (Basel) 2023; 16:1628. [PMID: 38004493 PMCID: PMC10675193 DOI: 10.3390/ph16111628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
In multiple myeloma impressive outcomes have improved with the introduction of new therapeutic approaches, mainly those including naked monoclonal antibodies such as daratumumab and isatuximab. However, moving to earlier lines of therapy with effective anti-myeloma drugs led to an increase in the number of patients who developed multi-refractoriness to them early on. Currently, triple- or multi-refractory MM represents an unmet medical need, and their management remains a complicated challenge. The recent approval of new immunotherapeutic approaches such as conjugated monoclonal antibodies, bispecific antibodies, and CAR T cells could be a turning point for these heavily pretreated patients. Nevertheless, several issues regarding their use are unsolved, such as how to select patients for each strategy or how to sequence these therapies within the MM therapeutic landscape. Here we provide an overview of the most recent data about approved conjugated monoclonal antibody belantamab, mafodotin, bispecific antibody teclistamab, and other promising compounds under development, mainly focusing on the ongoing clinical trials with monoclonal antibody combination approaches in advanced and earlier phases of MM treatment.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Laura Corvatta
- U.O.C. Medicina, Ospedale Profili, 60044 Fabriano, Italy
| | | | - Erika Morsia
- Clinica di Ematologia Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Antonella Poloni
- Clinica di Ematologia Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| | - Massimo Offidani
- Clinica di Ematologia Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy
| |
Collapse
|
25
|
van de Donk NWCJ, O'Neill C, de Ruijter MEM, Verkleij CPM, Zweegman S. T-cell redirecting bispecific and trispecific antibodies in multiple myeloma beyond BCMA. Curr Opin Oncol 2023; 35:601-611. [PMID: 37501530 PMCID: PMC10566598 DOI: 10.1097/cco.0000000000000983] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
PURPOSE OF REVIEW B-cell maturation antigen (BCMA)-directed T-cell immunotherapies, such as chimeric antigen receptor T-cells (CAR T-cells) and bispecific antibodies (BsAbs) have markedly improved the survival of triple-class refractory multiple myeloma (MM). However, the majority of patients still develops disease progression, underlining the need for new agents for these patients. RECENT FINDINGS Novel T-cell redirecting BsAbs targeting alternative tumor-associated antigens have shown great promise in heavily pretreated MM, including patients previously exposed to BCMA-directed therapies. This includes the G-protein-coupled receptor class 5 member D (GPRC5D)-targeting BsAbs talquetamab and forimtamig, as well as the Fc receptor-homolog 5 (FcRH5)-targeting BsAb cevostamab. Toxicity associated with these BsAbs includes cytokine-release syndrome, cytopenias, and infections. In addition, GPRC5D-targeting BsAbs are associated with specific 'on target/off tumor' toxicities including rash, nail disorders, and dysgeusia. Trispecifc antibodies targeting two different MM-associated antigens to prevent antigen escape are in early clinical development, as well as trispecific antibodies (TsAbs) that provide an additional co-stimulatory signal to T-cells to prevent their exhaustion. SUMMARY Various T-cell redirecting BsAbs are in advanced stages of clinical development with promising activity and a manageable toxicity profile. Ongoing studies are evaluating combination strategies, fixed-duration treatment, and use of BsAbs in earlier lines of therapy. TsAbs hold great promise for the future.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Chloe O'Neill
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Maaike E M de Ruijter
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
26
|
van de Donk NWCJ, Minnema MC, van der Holt B, Schjesvold F, Wu KL, Broijl A, Roeloffzen WWH, Gadisseur A, Pietrantuono G, Pour L, van der Velden VHJ, Lund T, Offidani M, Grasso M, Giaccone L, Razawy W, Tacchetti P, Mancuso K, Silkjaer T, Caers J, Zweegman S, Hájek R, Benjamin R, Vangsted AJ, Boccadoro M, Gay F, Sonneveld P, Musto P. Treatment of primary plasma cell leukaemia with carfilzomib and lenalidomide-based therapy (EMN12/HOVON-129): final analysis of a non-randomised, multicentre, phase 2 study. Lancet Oncol 2023; 24:1119-1133. [PMID: 37717583 DOI: 10.1016/s1470-2045(23)00405-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Primary plasma cell leukaemia is a rare and aggressive plasma cell disorder with a poor prognosis. The aim of the EMN12/HOVON-129 study was to improve the outcomes of patients with primary plasma cell leukaemia by incorporating carfilzomib and lenalidomide in induction, consolidation, and maintenance therapy. METHODS The EMN12/HOVON-129 study is a non-randomised, phase 2, multicentre study conducted at 19 academic centres and hospitals in seven European countries (Belgium, Czech Republic, Denmark, Italy, Norway, The Netherlands, and the UK) for previously untreated patients with primary plasma cell leukaemia aged 18 years or older. Inclusion criteria were newly diagnosed primary plasma cell leukaemia (defined as >2 ×109 cells per L circulating monoclonal plasma cells or plasmacytosis >20% of the differential white cell count) and WHO performance status 0-3. Patients aged 18-65 years (younger patients) and 66 years or older (older patients) were treated in age-specific cohorts and were analysed separately. Younger patients were treated with four 28-day cycles of carfilzomib (36 mg/m2 intravenously on days 1, 2, 8, 9, 15, and 16), lenalidomide (25 mg orally on days 1-21), and dexamethasone (20 mg orally on days 1, 2, 8, 9, 15, 16, 22, and 23). Carfilzomib-lenalidomide-dexamethasone (KRd) induction was followed by double autologous haematopoietic stem-cell transplantation (HSCT), four cycles of KRd consolidation, and then maintenance with carfilzomib (27 mg/m2 intravenously on days 1, 2, 15, and 16 for the first 12 28-day cycles, and then 56 mg/m2 on days 1 and 15 in all subsequent cycles) and lenalidomide (10 mg orally on days 1-21) until progression. Patients who were eligible for allogeneic HSCT, could also receive a single autologous HSCT followed by reduced-intensity conditioning allogeneic HSCT and then carfilzomib-lenalidomide maintenance. Older patients received eight cycles of KRd induction followed by maintenance therapy with carfilzomib and lenalidomide until progression. The primary endpoint was progression-free survival. The primary analysis population was the intention-to-treat population, irrespective of the actual treatment received. Data from all participants who received any study drug were included in the safety analyses. The trial was registered at www.trialregister.nl (until June 2022) and https://trialsearch.who.int/ as NTR5350; recruitment is complete and this is the final analysis. FINDINGS Between Oct 23, 2015, and Aug 5, 2021, 61 patients were enrolled and received KRd induction treatment (36 patients aged 18-65 years [20 (56%) were male and 16 (44%) female], and 25 aged ≥66 years [12 (48%) were male and 13 (52%) female]). With a median follow-up of 43·5 months (IQR 27·7-67·8), the median progression-free survival was 15·5 months (95% CI 9·4-38·4) for younger patients. For older patients, median follow-up was 32·0 months (IQR 24·7-34·6), and median progression-free survival was 13·8 months (95% CI 9·2-35·5). Adverse events were most frequently observed directly after treatment initiation, with infections (two of 36 (6%) younger patients and eight of 25 (32%) older patients) and respiratory events (two of 36 [6%] younger patients and four of 25 [16%] older patients) being the most common grade 3 or greater events during the first four KRd cycles. Treatment-related serious adverse events were reported in 26 (72%) of 36 younger patients and in 19 (76%) of 25 older patients, with infections being the most common. Treatment-related deaths were reported in none of the younger patients and three (12%) of the older patients (two infections and one unknown cause of death). INTERPRETATION Carfilzomib and lenalidomide-based therapy provides improved progression-free survival compared with previously published data. However, results remain inferior in primary plasma cell leukaemia compared with multiple myeloma, highlighting the need for new studies incorporating novel immunotherapies. FUNDING Dutch Cancer Society, Celgene (a BMS company), and AMGEN.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands.
| | - Monique C Minnema
- University Medical Center Utrecht, Department of Hematology, Utrecht University, Utrecht, Netherlands
| | - Bronno van der Holt
- HOVON Foundation, Rotterdam, Netherlands; Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Fredrik Schjesvold
- Oslo Myeloma Center, Department of Hematology, Oslo University Hospital and KG Jebsen Center for B cell malignancies, University of Oslo, Oslo, Norway
| | - Ka Lung Wu
- Department of Hematology, ZNA Stuivenberg, Antwerp, Belgium
| | - Annemiek Broijl
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Wilfried W H Roeloffzen
- Department of Hematology, University Medical Center Groningen, University Groningen, Groningen, Netherlands
| | - Alain Gadisseur
- Department of Haematology, Antwerp University Hospital, Edegem, Belgium
| | - Giuseppe Pietrantuono
- Unit of Hematology, Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | - Ludek Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, Brno, Czech Republic
| | | | | | | | | | - Luisa Giaccone
- Department of Oncology and Hematology, SSD Stem Cell Transplant Center, AOU Citta della Salute e della Scienza di Torino, Torino, Italy
| | | | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematolgia Seràgnoli, Bologna, Italy
| | - Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematolgia Seràgnoli, Bologna, Italy; Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | | | - Jo Caers
- Department of Hematology, CHU Liege, Liege, Belgium
| | - Sonja Zweegman
- Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Roman Hájek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic; Department of Hematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | | | - Annette Juul Vangsted
- Department of Hematology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | | | - Francesca Gay
- Division of Hematology, Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Pellegrino Musto
- Department of Precision and Regenerative Medicine and Ionian Area, Aldo Moro University School of Medicine, and Hematology and Stem Cell Transplantation Unit, AOU Consorziale Policlinico, Bari, Italy
| |
Collapse
|
27
|
Hussain M, Yellapragada S, Al Hadidi S. Differential Diagnosis and Therapeutic Advances in Multiple Myeloma: A Review Article. Blood Lymphat Cancer 2023; 13:33-57. [PMID: 37731771 PMCID: PMC10508231 DOI: 10.2147/blctt.s272703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by the abnormal clonal proliferation of plasma cells that may result in focal bone lesions, renal failure, anemia, and/or hypercalcemia. Recently, the diagnosis and treatment of MM have evolved due to a better understanding of disease pathophysiology, improved risk stratification, and new treatments. The incorporation of new drugs, including proteasome inhibitors, immunomodulatory drugs, anti-CD38 antibodies and high-dose chemotherapy followed by hematopoietic stem cell transplantation, has resulted in a significant improvement in patient outcomes and QoL. In this review, we summarize differential diagnoses and therapeutic advances in MM.
Collapse
Affiliation(s)
- Munawwar Hussain
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sarvari Yellapragada
- Michael E. DeBakey VA Medical Center and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Samer Al Hadidi
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
28
|
O'Neill C, van de Donk NWCJ. T-cell redirecting bispecific antibodies in multiple myeloma: Current landscape and future directions. EJHAEM 2023; 4:811-822. [PMID: 37601851 PMCID: PMC10435697 DOI: 10.1002/jha2.729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/25/2023] [Indexed: 08/22/2023]
Abstract
T-cell engaging bispecific antibodies (BsAbs) have substantial activity in heavily pretreated patients with multiple myeloma (MM). The overall response rate obtained with B-cell maturation antigen (BCMA)-targeting BsAbs is approximately 60%-70%, including a high proportion of patients achieving very good partial response or complete response. Comparable efficacy is seen with BsAbs targeting GPRC5D or FcRH5. Cytokine release syndrome is frequently observed with BsAb treatment, but mostly during the step-up doses and the first full dose. Early intervention with IL-6 receptor blocking antibodies (e.g., tocilizumab) prevents escalation to severe manifestations. Infections are also common during treatment and related to the extent of exposure to immune suppressive anti-MM agents, as well as development of hypogammaglobulinemia due to elimination of normal plasma cells, and probably because of T-cell exhaustion resulting from continuous BsAb-mediated T-cell activation. Adequate monitoring for infections and institution of infectious prophylaxis are essential. Patients treated with GPRC5D-targteing BsAbs often develop skin and nail disorders and loss of taste, which is likely related to GPRC5D expression in cells that produce hard keratin. Currently ongoing studies are aiming at further improving these results by evaluating BsAbs in combination with other drugs, such as immunomodulatory agents and anti-CD38 antibodies, as well as the application of BsAbs in earlier lines of therapy, including patients with newly diagnosed disease. We expect that the outcomes of patients with MM will further improve by the introduction of this novel type of T-cell immunotherapy.
Collapse
Affiliation(s)
- Chloe O'Neill
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of HematologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Biology and ImmunologyAmsterdamThe Netherlands
| | - Niels W. C. J. van de Donk
- Amsterdam UMC, Vrije Universiteit AmsterdamDepartment of HematologyAmsterdamThe Netherlands
- Cancer Center AmsterdamCancer Biology and ImmunologyAmsterdamThe Netherlands
| |
Collapse
|
29
|
van de Donk NWCJ, Zweegman S. T-cell-engaging bispecific antibodies in cancer. Lancet 2023; 402:142-158. [PMID: 37271153 DOI: 10.1016/s0140-6736(23)00521-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/06/2023] [Accepted: 03/02/2023] [Indexed: 06/06/2023]
Abstract
T-cell-engaging bispecific antibodies (BsAbs) simultaneously bind to antigens on tumour cells and CD3 subunits on T cells. This simultaneous binding results in the recruitment of T cells to the tumour, followed by T-cell activation and degranulation, and tumour cell elimination. T-cell-engaging BsAbs have shown substantial activity in several haematological malignancies by targeting CD19 in acute lymphoblastic leukaemia, CD20 in B-cell non-Hodgkin lymphoma, and BCMA and GPRC5D in multiple myeloma. Progress with solid tumours has been slower, in part due to the paucity of therapeutic targets with a tumour-specific expression profile, which is needed to limit on-target off-tumour side-effects. Nevertheless, BsAb-mediated recognition of a peptide fragment of gp100 presented by HLA-A2:01 molecules has shown marked activity in patients with unresectable or metastatic uveal melanoma. Cytokine release syndrome is the most frequent toxicity associated with BsAb treatment and is caused by activated T cells secreting proinflammatory cytokines. Understanding of resistance mechanisms has resulted in the development of new T cell-redirecting formats and novel combination strategies, which are expected to further improve depth and duration of response.
Collapse
Affiliation(s)
- Niels W C J van de Donk
- Amsterdam University Medical Centres, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands.
| | - Sonja Zweegman
- Amsterdam University Medical Centres, Vrije Universiteit Amsterdam, Department of Hematology, Amsterdam, Netherlands; Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
30
|
Grosicki S, Bednarczyk M, Kociszewska K. Elranatamab: a new promising BispAb in multiple myeloma treatment. Expert Rev Anticancer Ther 2023; 23:775-782. [PMID: 37434334 DOI: 10.1080/14737140.2023.2236303] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/10/2023] [Indexed: 07/13/2023]
Abstract
INTRODUCTION Multiple myeloma is a B-cell malignancy caused by proliferating plasma cells in the bone marrow microenvironment in collaboration with various cell lineage subsets and growth factors without any perfect regulation and tendency to clonal heterogeneity. Despite remarkable improvement in MM treatment and the overall survival of patients, multiple myeloma remains an incurable disease with the tendency to relapse. Therefore, there is an urgent need for new therapeutic options to provide a stabilized response to treatment with long-term duration. AREAS COVERED Elranatamab (PF-06863135), is a novel heterodimeric humanized full-length bispecific IgG2 kappa antibody derived from 2 mAbs, the anti-BCMA mAb (PF-06863058) and the anti-CD3 mAb (PF-06863059), not yet licensed in routine use. This binding affinity of elranatamab to BCMA and CD3 has been optimized to potentially prompt more potent T cell-mediated anti-myeloma activity. Subcutaneous (s.c.) administration of elranatamab is superior in comparison to intravenous (i.v.), thus it is associated with lower incidence of adverse events, even in higher doses. EXPERT OPINION Currently, elranatamab is being investigated in a few clinical studies, and the preliminary results are very encouraging. At the time of writing this review there were no full papers published and all of the data in the literature were based on abstract presentations which carry limitations.
Collapse
Affiliation(s)
- Sebastian Grosicki
- Medical University of Silesia, Department of Hematology and Cancer Prevention, Katowice, Slaskie, Poland
| | - Martyna Bednarczyk
- Medical University of Silesia, Department of Hematology and Cancer Prevention, Katowice, Slaskie, Poland
| | - Karolina Kociszewska
- Medical University of Silesia, Department of Hematology and Cancer Prevention, Katowice, Slaskie, Poland
| |
Collapse
|
31
|
Hiemstra IH, Santegoets KCM, Janmaat ML, De Goeij BECG, Ten Hagen W, van Dooremalen S, Boross P, van den Brakel J, Bosgra S, Andringa G, van Kessel-Welmers B, Verzijl D, Hibbert RG, Frerichs KA, Mutis T, van de Donk NWCJ, Ahmadi T, Satijn D, Sasser AK, Breij ECW. Preclinical anti-tumour activity of HexaBody-CD38, a next-generation CD38 antibody with superior complement-dependent cytotoxic activity. EBioMedicine 2023; 93:104663. [PMID: 37379657 DOI: 10.1016/j.ebiom.2023.104663] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
BACKGROUND HexaBody®-CD38 (GEN3014) is a hexamerization-enhanced human IgG1 that binds CD38 with high affinity. The E430G mutation in its Fc domain facilitates the natural process of antibody hexamer formation upon binding to the cell surface, resulting in increased binding of C1q and potentiated complement-dependent cytotoxicity (CDC). METHODS Co-crystallization studies were performed to identify the binding interface of HexaBody-CD38 and CD38. HexaBody-CD38-induced CDC, antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), trogocytosis, and apoptosis were assessed using flow cytometry assays using tumour cell lines, and MM patient samples (CDC). CD38 enzymatic activity was measured using fluorescence spectroscopy. Anti-tumour activity of HexaBody-CD38 was assessed in patient-derived xenograft mouse models in vivo. FINDINGS HexaBody-CD38 binds a unique epitope on CD38 and induced potent CDC in multiple myeloma (MM), acute myeloid leukaemia (AML), and B-cell non-Hodgkin lymphoma (B-NHL) cells. Anti-tumour activity was confirmed in patient-derived xenograft models in vivo. Sensitivity to HexaBody-CD38 correlated with CD38 expression level and was inversely correlated with expression of complement regulatory proteins. Compared to daratumumab, HexaBody-CD38 showed enhanced CDC in cell lines with lower levels of CD38 expression, without increasing lysis of healthy leukocytes. More effective CDC was also confirmed in primary MM cells. Furthermore, HexaBody-CD38 efficiently induced ADCC, ADCP, trogocytosis, and apoptosis after Fc-crosslinking. Moreover, HexaBody-CD38 strongly inhibited CD38 cyclase activity, which is hypothesized to relieve immune suppression in the tumour microenvironment. INTERPRETATION Based on these preclinical studies, a clinical trial was initiated to assess the clinical safety of HexaBody-CD38 in patients with MM. FUNDING Genmab.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kristine A Frerichs
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Tuna Mutis
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
32
|
Terpos E, Neri P, van de Donk NWCJ, D'Agostino M, Parekh S, Jagannath S, Ludwig H, Avigan DE, Dhodapkar MV, Raje NS. Immune Reconstitution and Vaccinations in Multiple Myeloma: A Report From the 19th International Myeloma Society Annual Workshop. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:413-419. [PMID: 37055346 DOI: 10.1016/j.clml.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
Given the significance of the immune system and the important role of therapies within the context of the immune system in plasma cell disorders, the International Myeloma Society annual workshop convened a session dedicated to this topic. A panel of experts covered various aspects of immune reconstitution and vaccination. The top oral presentations were highlighted and discussed. This is a report of the proceedings.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Paola Neri
- Department of Medical Oncology and Hematology, Tom Baker Cancer Center, Calgary, Alberta, Canada; Arnie Charbonneau Cancer Research Institute, Calgary, Alberta, Canada
| | - Niels W C J van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mattia D'Agostino
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Samir Parekh
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sundar Jagannath
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Heinz Ludwig
- Department of Medicine I, Center for Medical Oncology and Hematology with Outpatient Department and Palliative Care, Wilhelminen Cancer Research Institute, Vienna, Austria
| | - David E Avigan
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Madhav V Dhodapkar
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA
| | - Noopur S Raje
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA; Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
33
|
Friedrich MJ, Neri P, Kehl N, Michel J, Steiger S, Kilian M, Leblay N, Maity R, Sankowski R, Lee H, Barakat E, Ahn S, Weinhold N, Rippe K, Bunse L, Platten M, Goldschmidt H, Müller-Tidow C, Raab MS, Bahlis NJ. The pre-existing T cell landscape determines the response to bispecific T cell engagers in multiple myeloma patients. Cancer Cell 2023; 41:711-725.e6. [PMID: 36898378 DOI: 10.1016/j.ccell.2023.02.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 11/02/2022] [Accepted: 02/08/2023] [Indexed: 03/11/2023]
Abstract
Bispecific T cell engagers (TCEs) have shown promise in the treatment of various cancers, but the immunological mechanism and molecular determinants of primary and acquired resistance to TCEs remain poorly understood. Here, we identify conserved behaviors of bone marrow-residing T cells in multiple myeloma patients undergoing BCMAxCD3 TCE therapy. We show that the immune repertoire reacts to TCE therapy with cell state-dependent clonal expansion and find evidence supporting the coupling of tumor recognition via major histocompatibility complex class I (MHC class I), exhaustion, and clinical response. We find the abundance of exhausted-like CD8+ T cell clones to be associated with clinical response failure, and we describe loss of target epitope and MHC class I as tumor-intrinsic adaptations to TCEs. These findings advance our understanding of the in vivo mechanism of TCE treatment in humans and provide the rationale for predictive immune-monitoring and conditioning of the immune repertoire to guide future immunotherapy in hematological malignancies.
Collapse
Affiliation(s)
- Mirco J Friedrich
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Paola Neri
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Niklas Kehl
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julius Michel
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Simon Steiger
- Division of Chromatin Networks, BioQuant Center & German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Michael Kilian
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Noémie Leblay
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Ranjan Maity
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Roman Sankowski
- Department of Neuropathology, Freiburg University Hospital, Freiburg, Germany
| | - Holly Lee
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Elie Barakat
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Sungwoo Ahn
- Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada
| | - Niels Weinhold
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, BioQuant Center & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lukas Bunse
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Platten
- Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Neurology, MCTN, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Helmholtz Institute of Translational Oncology (HI-TRON), Mainz, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim Germany
| | - Hartmut Goldschmidt
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Marc-Steffen Raab
- Department of Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany; Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Nizar J Bahlis
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Canada; Tom Baker Cancer Center, Department of Hematology and Oncology, Calgary, Canada.
| |
Collapse
|
34
|
Varughese P, Smith R, Xue M, Dorrow N, Hogea C, Maiese EM, Buckingham T. Real-world treatment patterns and outcomes of triple-class treated patients with multiple myeloma in the United States. Expert Rev Hematol 2023; 16:65-74. [PMID: 36579455 DOI: 10.1080/17474086.2023.2154648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Although multiple myeloma (MM) survival has improved following the introduction of proteosome inhibitors, immunomodulatory drugs, and anti-CD38 therapies, patients become refractory to these agents. Real-world outcomes of triple-class exposed patients are limited and were investigated in this study. METHODS The Integra Connect Database was used to assess the treatment patterns of triple-class exposed patients with relapsed/refractory MM (RRMM) (January 2016-December 2019). RESULTS During this period, patients (N = 501) reached triple exposure in a median of three lines of therapy (LOTs) over 995 days. A new LOT was started in a median of 18 (1-691) days after triple exposure; 71% of the patients started a new LOT within 30 days. Throughout the follow-up period, 8% of the patients had a therapy gap greater than 90 days. Following triple exposure, 103/501 patients (21%) received only triple-class agents in subsequent LOTs, while 24 (4.8%) patients received only non-triple-class agents. The median apparent survival from initiation of first therapy after triple exposure was 308 days. CONCLUSION These results indicate that recycling of triple-class agents after previous exposure is widespread and prognosis in the RRMM population remains poor, highlighting the continuing unmet need for new agents with novel mechanisms to improve patient outcomes.
Collapse
Affiliation(s)
| | | | - Mei Xue
- IntegraConnect, West Palm Beach, FL, USA
| | | | | | | | | |
Collapse
|
35
|
Banerjee R, Lee SS, Cowan AJ. Innovation in BCMA CAR-T therapy: Building beyond the Model T. Front Oncol 2022; 12:1070353. [PMID: 36505779 PMCID: PMC9729952 DOI: 10.3389/fonc.2022.1070353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Autologous chimeric antigen receptor T-cell (CAR-T) therapies targeting B-cell maturation antigen (BCMA) have revolutionized the field of multiple myeloma in the same way that the Ford Model T revolutionized the original CAR world a century ago. However, we are only beginning to understand how to improve the efficacy and usability of these cellular therapies. In this review, we explore three automotive analogies for innovation with BCMA CAR-T therapies: stronger engines, better mileage, and hassle-free delivery. Firstly, we can build stronger engines in terms of BCMA targeting: improved antigen binding, tools to modulate antigen density, and armoring to better reach the antigen itself. Secondly, we can improve "mileage" in terms of response durability through ex vivo CAR design and in vivo immune manipulation. Thirdly, we can implement hassle-free delivery through rapid manufacturing protocols and off-the-shelf products. Just as the Model T set a benchmark for car manufacturing over 100 years ago, idecabtagene vicleucel and ciltacabtagene autoleucel have now set the starting point for BCMA CAR-T therapy with their approvals. As with any emerging technology, whether automotive or cellular, the best in innovation and optimization is yet to come.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Andrew J. Cowan
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
36
|
Cho SF, Yeh TJ, Anderson KC, Tai YT. Bispecific antibodies in multiple myeloma treatment: A journey in progress. Front Oncol 2022; 12:1032775. [PMID: 36330495 PMCID: PMC9623099 DOI: 10.3389/fonc.2022.1032775] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 07/29/2023] Open
Abstract
The incorporation of novel agents and monoclonal antibody-based therapies into the treatment of multiple myeloma (MM) has significantly improved long-term patient survival. However, the disease is still largely incurable, with high-risk patients suffering shorter survival times, partly due to weakened immune systems. Bispecific molecules, including bispecific antibodies (BisAbs) and bispecific T-cell engagers (BiTEs), encourage immune cells to lyse MM cells by simultaneously binding antigens on MM cells and immune effector cells, bringing those cells into close proximity. BisAbs that target B-cell maturation antigen (BCMA) and GPRC5D have shown impressive clinical activity, and the results of early-phase clinical trials targeting FcRH5 in patients with relapsed/refractory MM (RRMM) are also promising. Furthermore, the safety profile of these agents is favorable, including mainly low-grade cytokine release syndrome (CRS). These off-the-shelf bispecific molecules will likely become an essential part of the MM treatment paradigm. Here, we summarize and highlight various bispecific immunotherapies under development in MM treatment, as well as the utility of combining them with current standard-of-care treatments and new strategies. With the advancement of novel combination treatment approaches, these bispecific molecules may lead the way to a cure for MM.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Jang Yeh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kenneth C. Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
37
|
Holstein SA, Asimakopoulos F, Azab AK, Bianchi G, Bhutani M, Crews LA, Cupedo T, Giles H, Gooding S, Hillengass J, John L, Kaiser S, Lee L, Maclachlan K, Pasquini MC, Pichiorri F, Shah N, Shokeen M, Shy BR, Smith EL, Verona R, Usmani SZ, McCarthy PL. Proceedings from the Blood and Marrow Transplant Clinical Trials Network Myeloma Intergroup Workshop on Immune and Cellular Therapy in Multiple Myeloma. Transplant Cell Ther 2022; 28:446-454. [PMID: 35605882 PMCID: PMC9357156 DOI: 10.1016/j.jtct.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022]
Abstract
The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) Myeloma Intergroup conducted a workshop on Immune and Cellular Therapy in Multiple Myeloma on January 7, 2022. This workshop included presentations by basic, translational, and clinical researchers with expertise in plasma cell dyscrasias. Four main topics were discussed: platforms for myeloma disease evaluation, insights into pathophysiology, therapeutic target and resistance mechanisms, and cellular therapy for multiple myeloma. Here we provide a comprehensive summary of these workshop presentations.
Collapse
Affiliation(s)
| | - Fotis Asimakopoulos
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - Giada Bianchi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Leslie A Crews
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Tom Cupedo
- ErasmusMC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Hannah Giles
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Sarah Gooding
- MRC Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Lukas John
- University Hospital Heidelberg, Heidelberg, Germany
| | | | - Lydia Lee
- University College London, London, United Kingdom
| | | | | | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California; Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, California
| | - Nina Shah
- University of California San Francisco, San Francisco, California
| | - Monica Shokeen
- Washington University School of Medicine, St. Louis, Missouri
| | - Brian R Shy
- University of California San Francisco, San Francisco, California
| | - Eric L Smith
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Raluca Verona
- Janssen Research & Development, Spring House, Pennsylvania
| | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
38
|
Translational Modeling Predicts Efficacious Therapeutic Dosing Range of Teclistamab for Multiple Myeloma. Target Oncol 2022; 17:433-439. [PMID: 35749004 PMCID: PMC9345835 DOI: 10.1007/s11523-022-00893-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 10/25/2022]
Abstract
BACKGROUND Teclistamab (JNJ-64007957), a B-cell maturation antigen × CD3 bispecific antibody, displayed potent T-cell-mediated cytotoxicity of multiple myeloma cells in preclinical studies. OBJECTIVE A first-in-human, Phase I, dose escalation study (MajesTEC-1) is evaluating teclistamab in patients with relapsed/refractory multiple myeloma. PATIENTS AND METHODS To estimate the efficacious therapeutic dosing range of teclistamab, pharmacokinetic (PK) data following the first cycle doses in the low-dose cohorts in the Phase I study were modeled using a 2-compartment model and simulated to predict the doses that would have average and trough serum teclistamab concentrations in the expected therapeutic range (between EC50 and EC90 values from an ex vivo cytotoxicity assay). RESULTS The doses predicted to have average serum concentrations between the EC50 and EC90 range were validated. In addition, simulations showed that weekly intravenous and subcutaneous doses of 0.70 mg/kg and 0.72 mg/kg, respectively, resulted in mean trough levels comparable to the maximum EC90. The most active doses in the Phase I study were weekly intravenous doses of 0.27 and 0.72 mg/kg and weekly subcutaneous doses of 0.72 and 1.5 mg/kg, with the weekly 1.5 mg/kg subcutaneous doses selected as the recommended Phase II dose (RP2D). With active doses, exposure was maintained above the mean EC90. All patients who responded to the RP2D of teclistamab had exposure above the maximum EC90 in both serum and bone marrow on cycle 3, Day 1 of treatment. CONCLUSIONS Our findings show that PK simulations of early clinical data together with ex vivo cytotoxicity estimates can inform the identification of a bispecific antibody's therapeutic range. CLINICAL TRIAL REGISTRATION NCT03145181, date of registration: May 9, 2017.
Collapse
|
39
|
Martens AWJ, Rietveld JM, de Boer R, Peters FS, Ngo A, van Mil LWHG, de Heer K, Spaargaren M, Verkleij CPM, van de Donk NWCJ, Adams HC, Eldering E, van Noesel CJM, Verona R, Kater AP. Redirecting T-cell Activity with Anti-BCMA/Anti-CD3 Bispecific Antibodies in Chronic Lymphocytic Leukemia and Other B-cell Lymphomas. CANCER RESEARCH COMMUNICATIONS 2022; 2:330-341. [PMID: 36875718 PMCID: PMC9981202 DOI: 10.1158/2767-9764.crc-22-0083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022]
Abstract
T-cell redirecting bispecific antibodies hold high promise for treatment of B-cell malignancies. B-cell maturation antigen (BCMA) exhibits high expression on normal and malignant mature B cells including plasma cells, which can be enhanced by inhibition of γ-secretase. BCMA is considered a validated target in multiple myeloma but whether mature B-cell lymphomas can be targeted by the BCMAxCD3 T-cell redirector teclistamab is currently unknown. BCMA expression on B-cell non-Hodgkin lymphoma and primary chronic lymphocytic leukemia (CLL) cells was assessed by flow cytometry and/or IHC. To assess teclistamab efficacy, cells were treated with teclistamab in presence of effector cells with/without γ-secretase inhibition. BCMA could be detected on all tested mature B-cell malignancy cell lines, while expression levels varied per tumor type. γ-secretase inhibition universally increased BCMA surface expression. These data were corroborated in primary samples from patients with Waldenstrom's macroglobulinemia, CLL, and diffuse large B-cell lymphoma. Functional studies with the B-cell lymphoma cell lines revealed teclistamab-mediated T-cell activation, proliferation, and cytotoxicity. This was independent of the level of BCMA expression, but generally lower in mature B-cell malignancies compared with multiple myeloma. Despite low BCMA levels, healthy donor T cells and CLL-derived T cells induced lysis of (autologous) CLL cells upon addition of teclistamab. These data show that BCMA is expressed on various B-cell malignancies and that lymphoma cell lines and primary CLL can be targeted using teclistamab. Further studies to understand the determinants of response to teclistamab are required to identify which other diseases might be suitable for teclistamab targeting. Significance Besides reported BCMA expression on multiple myeloma, we demonstrate BCMA can be detected and enhanced using γ-secretase inhibition on cell lines and primary material of various B-cell malignancies. Furthermore, using CLL we demonstrate that low BCMA-expressing tumors can be targeted efficiently using the BCMAxCD3 DuoBody teclistamab.
Collapse
Affiliation(s)
- Anne W J Martens
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam, the Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - Joanne M Rietveld
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Renate de Boer
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Fleur S Peters
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam, the Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands
| | - An Ngo
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lotte W H G van Mil
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Koen de Heer
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Hematology, Flevoziekenhuis, Almere, the Netherlands
| | - Marcel Spaargaren
- Cancer Center Amsterdam, Amsterdam, the Netherlands.,Department of Pathology, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, the Netherlands
| | - Christie P M Verkleij
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Homer C Adams
- Janssen Pharmaceutical Companies of Johnson & Johnson, Philadelphia, Pennsylvania
| | - Eric Eldering
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam, the Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, the Netherlands
| | - Carel J M van Noesel
- Department of Pathology, University of Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, the Netherlands
| | - Raluca Verona
- Janssen Pharmaceutical Companies of Johnson & Johnson, Philadelphia, Pennsylvania
| | - Arnon P Kater
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam, the Netherlands.,Amsterdam Infection & Immunity Institute, Amsterdam, the Netherlands.,Lymphoma and Myeloma Center Amsterdam, LYMMCARE, the Netherlands
| |
Collapse
|
40
|
T-cell redirecting bispecific antibodies in multiple myeloma: a revolution? Blood 2022; 139:3681-3687. [PMID: 35404996 DOI: 10.1182/blood.2021014611] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/01/2022] [Indexed: 11/20/2022] Open
Abstract
Bispecific antibodies are designed to link a surface target molecule on the malignant plasma cells to CD3 on T-cells and thereby redirect activated T-cells to induce tumor cell death. Early-phase clinical trials targeting B-cell maturation antigen, GPRC5D or FcRH5, have demonstrated a favorable safety profile and promising efficacy data in triple-class refractory multiple myeloma. This novel immunotherapeutic modality will likely change the treatment paradigm in the coming years.
Collapse
|
41
|
Tai W, Wahab A, Franco D, Shah Z, Ashraf A, Abid QUA, Mohammed YN, Lal D, Anwer F. Emerging Role of Antibody-Drug Conjugates and Bispecific Antibodies for the Treatment of Multiple Myeloma. Antibodies (Basel) 2022; 11:22. [PMID: 35466275 PMCID: PMC9036234 DOI: 10.3390/antib11020022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is characterized by malignant proliferation of malignant plasma cells; it is the second most common hematological malignancy associated with significant morbidity. Genetic intricacy, instability, and diverse clinical presentations remain a barrier to cure. The treatment of MM is modernized with the introduction of newer therapeutics agents, i.e., target-specific monoclonal antibodies. The currently available literature lacks the benefits of newer targeted therapy being developed with an aim to reduce side effects and increase effectiveness, compared to conventional chemotherapy regimens. This article aims to review literature about the current available monoclonal antibodies, antibody-drug conjugates, and bispecific antibodies for the treatment of MM.
Collapse
Affiliation(s)
- Waqqas Tai
- Department of Internal Medicine, The Brooklyn Hospital Center, Brooklyn, NY 11201, USA;
| | - Ahsan Wahab
- Department of Internal Medicine, Prattville Baptist Hospital, Prattville, AL 36066, USA;
| | - Diana Franco
- Department of Internal Medicine, Loyola MacNeal Hospital, Berwyn, IL 60402, USA;
| | - Zunairah Shah
- Department of Internal Medicine, Weiss Memorial Hospital, Chicago, IL 60640, USA;
| | - Aqsa Ashraf
- Department of Internal Medicine, Northwell Health, Mather Hospital, Port Jefferson, NY 11777, USA;
| | - Qurrat-Ul-Ain Abid
- Department of Internal Medicine, AMITA Health Saint Joseph Hospital, Chicago, IL 60657, USA;
| | - Yaqub Nadeem Mohammed
- Department of Internal Medicine, St. Joseph Mercy Oakland Hospital, Pontiac, MI 48341, USA;
| | - Darshan Lal
- Department of Internal Medicine, University of Nevada School of Medicine, Las Vegas, NV 89102, USA;
| | - Faiz Anwer
- Taussig Cancer Center, Myeloma Program, Cleveland Clinic, Cleveland, OH 44106, USA
| |
Collapse
|
42
|
Escure G, Manier S. [Bispecific antibodies in multiple myeloma]. Bull Cancer 2021; 108:S205-S212. [PMID: 34920804 DOI: 10.1016/j.bulcan.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 11/18/2022]
Abstract
Immunotherapies have recently emerged as potential game changers in the treatment of multiple myeloma (MM). Those include monoclonal antibodies (targeting CD38 or CS1), bispecific antibodies (BsAb, mainly targeting BCMA, GPRC5D or FcRH5), antibody-drug conjugate (mainly targeting BCMA) and CAR-T cells (mainly targeting BCMA). BsAb have the capacity to bind two different antigens, one at the tumor cell surface and one on T cells (CD3), recreating the immune synapse. In this article, we discuss the main clinical data on BsAb in MM, as well as their different constructs and the potential mechanism of resistance.
Collapse
Affiliation(s)
- Guillaume Escure
- CHU Lille, hôpital Huriez, service d'hématologie, 59000 Lille, France; Université de Lille, Unité CANTHER, Inserm UMR-S1277 & CNRS UMR9020, 59000 Lille, France
| | - Salomon Manier
- CHU Lille, hôpital Huriez, service d'hématologie, 59000 Lille, France; Université de Lille, Unité CANTHER, Inserm UMR-S1277 & CNRS UMR9020, 59000 Lille, France.
| |
Collapse
|
43
|
Katsarou A, Sjöstrand M, Naik J, Mansilla-Soto J, Kefala D, Kladis G, Nianias A, Ruiter R, Poels R, Sarkar I, Patankar YR, Merino E, Reijmers RM, Frerichs KA, Yuan H, de Bruijn J, Stroopinsky D, Avigan D, van de Donk NW, Zweegman S, Mutis T, Sadelain M, Groen RW, Themeli M. Combining a CAR and a chimeric costimulatory receptor enhances T cell sensitivity to low antigen density and promotes persistence. Sci Transl Med 2021; 13:eabh1962. [PMID: 34878825 PMCID: PMC9869449 DOI: 10.1126/scitranslmed.abh1962] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Despite the high remission rates achieved using T cells bearing a chimeric antigen receptor (CAR) against hematogical malignancies, there is still a considerable proportion of patients who eventually experience tumor relapse. Clinical studies have established that mechanisms of treatment failure include the down-regulation of target antigen expression and the limited persistence of effective CAR T cells. We hypothesized that dual targeting mediated by a CAR and a chimeric costimulatory receptor (CCR) could simultaneously enhance T cell cytotoxicity and improve durability. Concomitant high-affinity engagement of a CD38-binding CCR enhanced the cytotoxicity of BCMA-CAR and CD19-CAR T cells by increasing their functional binding avidity. In comparison to second-generation BCMA-CAR or CD19-CAR T cells, double-targeted CAR + CD38-CCR T cells exhibited increased sensitivity to recognize and lyse tumor variants of multiple myeloma and acute lymphoblastic leukemia with low antigen density in vitro. In addition, complimentary costimulation by 4-1BB and CD28 endodomains provided by the CAR and CCR combination conferred increased cytokine secretion and expansion and improved persistence in vivo. The cumulatively improved properties of CAR + CCR T cells enabled the in vivo eradication of antigen-low tumor clones, which were otherwise resistant to treatment with conventional CAR T cells. Therefore, multiplexing targeting and costimulation through the combination of a CAR and a CCR is a powerful strategy to improve the clinical outcomes of CAR T cells by enhancing cytotoxic efficacy and persistence, thus preventing relapses of tumor clones with low target antigen density.
Collapse
Affiliation(s)
- Afroditi Katsarou
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Maria Sjöstrand
- Center for Cell Engineering, Immunology Program, Memorial Sloan Kettering Cancer Center; NY 10065 New York, USA
| | - Jyoti Naik
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Jorge Mansilla-Soto
- Center for Cell Engineering, Immunology Program, Memorial Sloan Kettering Cancer Center; NY 10065 New York, USA
| | - Dionysia Kefala
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Georgios Kladis
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Alexandros Nianias
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Ruud Ruiter
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Renée Poels
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Irene Sarkar
- LUMICKS; Pilotenstraat 41 1059 CH Amsterdam, Netherlands
| | | | - Elena Merino
- LUMICKS; Pilotenstraat 41 1059 CH Amsterdam, Netherlands
| | | | - Kristine A. Frerichs
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Huipin Yuan
- Kuros Biosciences BV; 3723 MB Bilthoven, The Netherlands
| | - Joost de Bruijn
- Kuros Biosciences BV; 3723 MB Bilthoven, The Netherlands.,The School of Engineering and Materials Science, Queen Mary University of London; E1 4NS London, United Kingdom
| | - Dina Stroopinsky
- Beth Israel Deaconess Medical Center, Harvard Medical School; MA 02215 Boston, MA, USA
| | - David Avigan
- Beth Israel Deaconess Medical Center, Harvard Medical School; MA 02215 Boston, MA, USA
| | - Niels W.C.J. van de Donk
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Sonja Zweegman
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Tuna Mutis
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Michel Sadelain
- Center for Cell Engineering, Immunology Program, Memorial Sloan Kettering Cancer Center; NY 10065 New York, USA
| | - Richard W.J. Groen
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands
| | - Maria Themeli
- Department of Hematology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Cancer Center Amsterdam; 1081 HV Amsterdam, Netherlands.,Corresponding author: Maria Themeli MD PhD., VU University Medical Center, Dept. of Hematology, CCA 4.28, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands. Tel. +31 (0) 204447413,
| |
Collapse
|
44
|
Podar K, Leleu X. Relapsed/Refractory Multiple Myeloma in 2020/2021 and Beyond. Cancers (Basel) 2021; 13:5154. [PMID: 34680303 PMCID: PMC8534171 DOI: 10.3390/cancers13205154] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the challenges imposed by the COVID-19 pandemic, exciting therapeutic progress continues to be made in MM. New drug approvals for relapsed/refractory (RR)MM in 2020/2021 include the second CD38 monoclonal antibody, isatuximab, the first BCMA-targeting therapy and first-in-class antibody-drug conjugate (ADC) belantamab mafodotin, the first BCMA-targeting CAR T cell product Idecabtagen-Vicleucel (bb2121, Ide-Cel), the first in-class XPO-1 inhibitor selinexor, as well as the first-in-class anti-tumor peptide-drug conjugate, melflufen. The present introductory article of the Special Issue on "Advances in the Treatment of Relapsed and Refractory Multiple Myeloma: Novel Agents, Immunotherapies and Beyond" summarizes the most recent registration trials and emerging immunotherapies in RRMM, gives an overview on latest insights on MM genomics and on tumor-induced changes within the MM microenvironment, and presents some of the most promising rationally derived future therapeutic strategies.
Collapse
Affiliation(s)
- Klaus Podar
- Department of Internal Medicine 2, University Hospital Krems, Mitterweg 10, 3500 Krems an der Donau, Austria
- Molecular Oncology and Hematology Unit, Karl Landsteiner University of Health Sciences, Dr. Karl-Dorrek-Straße 30, 3500 Krems an der Donau, Austria
| | - Xavier Leleu
- Department of Hematology, and CIC1402 INSERM Unit, Poitiers University Hospital, 2 Rue de la Milétrie, 86021 Poitiers, France;
| |
Collapse
|
45
|
Hosny M, Verkleij CPM, van der Schans J, Frerichs KA, Mutis T, Zweegman S, van de Donk NWCJ. Current State of the Art and Prospects of T Cell-Redirecting Bispecific Antibodies in Multiple Myeloma. J Clin Med 2021; 10:4593. [PMID: 34640611 PMCID: PMC8509238 DOI: 10.3390/jcm10194593] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) patients eventually develop multi-drug-resistant disease with poor survival. Hence, the development of novel treatment strategies is of great importance. Recently, different classes of immunotherapeutic agents have shown great promise in heavily pre-treated MM, including T cell-redirecting bispecific antibodies (BsAbs). These BsAbs simultaneously interact with CD3 on effector T cells and a tumor-associated antigen on MM cells, resulting in redirection of T cells to MM cells. This leads to the formation of an immunologic synapse, the release of granzymes/perforins, and subsequent tumor cell lysis. Several ongoing phase 1 studies show substantial activity and a favorable toxicity profile with BCMA-, GPRC5D-, or FcRH5-targeting BsAbs in heavily pre-treated MM patients. Resistance mechanisms against BsAbs include tumor-related features, T cell characteristics, and impact of components of the immunosuppressive tumor microenvironment. Various clinical trials are currently evaluating combination therapy with a BsAb and another agent, such as a CD38-targeting antibody or an immunomodulatory drug (e.g., pomalidomide), to further improve response depth and duration. Additionally, the combination of two BsAbs, simultaneously targeting two different antigens to prevent antigen escape, is being explored in clinical studies. The evaluation of BsAbs in earlier lines of therapy, including newly diagnosed MM, is warranted, based on the efficacy of BsAbs in advanced MM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Niels W. C. J. van de Donk
- Cancer Center Amsterdam, Department of Hematology, Vrije Universiteit Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands; (M.H.); (C.P.M.V.); (J.v.d.S.); (K.A.F.); (T.M.); (S.Z.)
| |
Collapse
|
46
|
Mohan M, Maatman TC, Schinke C. The Role of Monoclonal Antibodies in the Era of Bi-Specifics Antibodies and CAR T Cell Therapy in Multiple Myeloma. Cancers (Basel) 2021; 13:4909. [PMID: 34638393 PMCID: PMC8507719 DOI: 10.3390/cancers13194909] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) remains largely incurable despite enormous improvement in the outcome of patients. Over the past decade, we have witnessed the "era of monoclonal antibody (moAb)", setting new benchmarks in clinical outcomes for relapsed and newly diagnosed MM. Due to their excellent efficacy and relative safe toxicity profile, moAbs in combination with immunomodulatory drugs (IMiDs) and proteasome inhibitors (PIs) have become the new backbone of upfront anti-MM therapy. Yet, most patients will eventually relapse and patients who become refractory to IMiDs, PIs and moAbs have a dismal outcome. Emerging T-cell directing therapies, such as bispecific antibody (bsAb) and chimeric antigen receptor T cells (CAR T) have shown unprecedented responses and outcomes in these heavily pretreated and treatment-refractory patients. Their clinical efficacy combined with high tolerability will likely lead to the use of these agents earlier in the treatment course and there is great enthusiasm that a combination of T cell directed therapy with moAbs can lead to long duration remission in the near future, possibly even without the need of high dose chemotherapy and stem cell transplantation. Herein, we summarize the role of naked moAbs in MM in the context of newer immunotherapeutic agents like bsAb and CAR T therapy.
Collapse
Affiliation(s)
- Meera Mohan
- Divicion of Hematology/Oncology, Froedtert Clinical Cancer Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA;
| | - Theresa Camille Maatman
- Divicion of Hematology/Oncology, Froedtert Clinical Cancer Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA;
| | - Carolina Schinke
- Myeloma Center, Division of Hematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
47
|
The Agony of Choice-Where to Place the Wave of BCMA-Targeted Therapies in the Multiple Myeloma Treatment Puzzle in 2022 and Beyond. Cancers (Basel) 2021; 13:cancers13184701. [PMID: 34572927 PMCID: PMC8471156 DOI: 10.3390/cancers13184701] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is no doubt that immunotherapeutic approaches will change the current treatment landscape of multiple myeloma in the near future; in particular, a wave of BCMA-targeted therapies is currently entering clinical routine. Although the increasing availability of different therapeutic approaches is highly welcome, it also increases the daily challenges in clinical decision making if they all use the same target. Here, we provide a comprehensive summary of BCMA-targeted approaches in myeloma and aim to share some basic concepts in clinical decision making. Abstract Since the introduction of first-generation proteasome inhibitors and immunomodulatory agents, the multiple myeloma (MM) treatment landscape has undergone a remarkable development. Most recently, immunotherapeutic strategies targeting the B cell maturation antigen (BCMA) entered the clinical stage providing access to highly anticipated novel treatment strategies. At present, numerous different approaches investigate BCMA as an effective multi-modal target. Currently, BCMA-directed antibody–drug conjugates, bispecific and trispecific antibodies, autologous and allogeneic CAR-T cell as well as CAR-NK cell constructs are either approved or in different stages of clinical and preclinical development for the treatment of MM. This armamentarium of treatment choices raises several challenges for clinical decision making, particularly in the absence of head-to-head comparisons. In this review, we provide a comprehensive overview of BCMA-targeting therapeutics, deliver latest updates on clinical trial data, and focus on potential patient selection criteria for different BCMA-targeting immunotherapeutic strategies.
Collapse
|
48
|
Krejcik J, Barnkob MB, Nyvold CG, Larsen TS, Barington T, Abildgaard N. Harnessing the Immune System to Fight Multiple Myeloma. Cancers (Basel) 2021; 13:4546. [PMID: 34572773 PMCID: PMC8467095 DOI: 10.3390/cancers13184546] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy differing substantially in clinical behavior, prognosis, and response to treatment. With the advent of novel therapies, many patients achieve long-lasting remissions, but some experience aggressive and treatment refractory relapses. So far, MM is considered incurable. Myeloma pathogenesis can broadly be explained by two interacting mechanisms, intraclonal evolution of cancer cells and development of an immunosuppressive tumor microenvironment. Failures in isotype class switching and somatic hypermutations result in the neoplastic transformation typical of MM and other B cell malignancies. Interestingly, although genetic alterations occur and evolve over time, they are also present in premalignant stages, which never progress to MM, suggesting that genetic mutations are necessary but not sufficient for myeloma transformation. Changes in composition and function of the immune cells are associated with loss of effective immune surveillance, which might represent another mechanism driving malignant transformation. During the last decade, the traditional view on myeloma treatment has changed dramatically. It is increasingly evident that treatment strategies solely based on targeting intrinsic properties of myeloma cells are insufficient. Lately, approaches that redirect the cells of the otherwise suppressed immune system to take control over myeloma have emerged. Evidence of utility of this principle was initially established by the observation of the graft-versus-myeloma effect in allogeneic stem cell-transplanted patients. A variety of new strategies to harness both innate and antigen-specific immunity against MM have recently been developed and intensively tested in clinical trials. This review aims to give readers a basic understanding of how the immune system can be engaged to treat MM, to summarize the main immunotherapeutic modalities, their current role in clinical care, and future prospects.
Collapse
Affiliation(s)
- Jakub Krejcik
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Charlotte Guldborg Nyvold
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, 5000 Odense, Denmark
| | - Thomas Stauffer Larsen
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Torben Barington
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Clinical Immunology, Odense University Hospital, 5000 Odense, Denmark
| | - Niels Abildgaard
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Odense University Hospital, 5000 Odense, Denmark; (J.K.); (M.B.B.); (C.G.N.); (T.S.L.); (T.B.)
- Department of Haematology, Odense University Hospital, 5000 Odense, Denmark
- Haematology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
49
|
Lancman G, Sastow DL, Cho HJ, Jagannath S, Madduri D, Parekh SS, Richard S, Richter J, Sanchez L, Chari A. Bispecific Antibodies in Multiple Myeloma: Present and Future. Blood Cancer Discov 2021; 2:423-433. [PMID: 34661161 PMCID: PMC8510808 DOI: 10.1158/2643-3230.bcd-21-0028] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
Despite many recent advances in therapy, there is still no plateau in overall survival curves in multiple myeloma. Bispecific antibodies are a novel immunotherapeutic approach designed to bind antigens on malignant plasma cells and cytotoxic immune effector cells. Early-phase clinical trials targeting B-cell maturation antigen (BCMA), GPRC5D, and FcRH5 have demonstrated a favorable safety profile, with mainly low-grade cytokine release syndrome, cytopenias, and infections. Although dose escalation is ongoing in several studies, early efficacy data show response rates in the most active dose cohorts between 61% and 83% with many deep responses; however, durability remains to be established. Further clinical trial data are eagerly anticipated. SIGNIFICANCE Overall survival of triple-class refractory multiple myeloma remains poor. Bispecific antibodies are a novel immunotherapeutic modality with a favorable safety profile and impressive preliminary efficacy in heavily treated patients. Although more data are needed, bispecifics will likely become an integral part of the multiple myeloma treatment paradigm in the near future. Studies in earlier lines of therapy and in combination with other active anti-multiple myeloma agents will help further define the role of bispecifics in multiple myeloma.
Collapse
Affiliation(s)
- Guido Lancman
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Hearn J Cho
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sundar Jagannath
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deepu Madduri
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Samir S Parekh
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Shambavi Richard
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joshua Richter
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Larysa Sanchez
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ajai Chari
- Tisch Cancer Insitute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Despite considerable advances in the treatment of multiple myeloma (MM) in the last decade, a significant number of patients still progress on current available therapies. Here, we review treatment modalities used to target BCMA in the treatment of MM, specifically antibody-drug conjugates (ADC), bispecific antibody constructs, and chimeric antibody receptor (CAR) modified T-cell therapies. We will provide an overview of therapies from these classes that have presented or published clinical data, as well as data on mechanisms of resistance to these novel agents. RECENT FINDINGS Clinical trials exploring different BCMA-targeting modalities to treat multiple myeloma are underway and demonstrate promising results. In relapsed/refractory multiple myeloma, anti-BCMA ADCs and bispecific antibody constructs are showing impressive efficacy with manageable side effect profiles. In parallel, adoptive cellular therapy has induced dramatic durable responses in multiply relapsed and refractory myeloma patients. Therapeutic approaches targeting BCMA hold significant potential in the management of multiple myeloma and will soon be incorporated in combination with current standard therapies to improve outcomes for patients with multiple myeloma. In addition, novel approaches are being evaluated to overcome resistance mechanisms to anti-BCMA therapies.
Collapse
Affiliation(s)
- Carlyn Rose Tan
- Myeloma Service Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 E 74th Street, New York, NY, 10021, USA.
| | - Urvi A Shah
- Myeloma Service Department of Medicine, Memorial Sloan Kettering Cancer Center, 530 E 74th Street, New York, NY, 10021, USA
| |
Collapse
|