1
|
Lu P, Zhang X, Yang J, Li J, Qiu L, Gong M, Wang H, Chen J, Liu H, Xiong M, Liu Y, Wang L. Nanobody-based naturally selected CD7-targeted CAR-T therapy for acute myeloid leukemia. Blood 2025; 145:1022-1033. [PMID: 39561281 DOI: 10.1182/blood.2024024861] [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: 04/01/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Approximately 30% of patients with acute myeloid leukemia (AML) express CD7 on their myeloblasts. We have previously demonstrated that single-chain variable fragment (scFv)-based "naturally selected" CD7 chimeric antigen receptor T-cell (NS7CAR-T) therapy shows significant efficacy, with a favorable safety profile in T-cell lymphoid malignancies. Here, we derived dual variable heavy-chain domain of a heavy-chain antibody (dVHH) NS7CAR-Ts that have superior CD7 binding specificity, affinity to their scFv-based counterparts, and improved proliferative capability. In this phase 1 clinical trial, we evaluated the efficacy and safety of nanobody-based dVHH NS7CAR-Ts for patients with CD7+ refractory/relapsed AML. A cohort of 10 patients received dVHH NS7CAR-Ts across 2 dosage levels of 5 × 105/kg and 1 × 106/kg. Before enrollment, patients had undergone a median of 8 (range, 3-17) prior lines of therapy. Seven patients had prior transplants. After NS7CAR-T infusion, 7 of 10 (70%) patients achieved complete remission (CR). The median observation time was 178 days (range, 28-776). Among 7 patients who achieved CR, 3 who relapsed from prior transplants underwent a second allogeneic hematopoietic stem cell transplant (allo-HSCT). One patient remained leukemia free on day 401, and the other 2 died on day 241 and day 776, respectively, from nonrelapse-related causes. Three CR patients without consolidative (allo-HSCT) relapsed within 90 days. All the nonresponders and relapsed patients had CD7 loss. The treatment was well tolerated, with 80% experiencing mild cytokine release syndrome and none had neurotoxicity. This trial underscores the potential promising treatment of dVHH NS7CAR-Ts in providing clinical benefits with a manageable safety profile to patients with CD7+ AML, warranting further investigation. This trial was registered at www.clinicaltrials.gov as #NCT04938115.
Collapse
Affiliation(s)
- Peihua Lu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Xian Zhang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Junfang Yang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Jingjing Li
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Liyuan Qiu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Meiwei Gong
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Hui Wang
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Jiaqi Chen
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Hongxing Liu
- Hebei Yanda Lu Daopei Hospital, Langfang, China
- Lu Daopei Institute of Hematology, Beijing, China
| | - Min Xiong
- Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Ying Liu
- Hebei Senlang Biotechnology Co, Ltd, Shijiazhuang, China
| | - Lin Wang
- Hebei Senlang Biotechnology Co, Ltd, Shijiazhuang, China
| |
Collapse
|
2
|
Suvannasankha A, Bahlis N, Trudel S, Weisel K, Koenecke C, Oriol A, Voorhees PM, Alonso AA, Callander NS, Mateos MV, Reddy N, Hakim S, LaMacchia J, Patel N, Williams D, Jewell RC, Zhou X, Gupta I, Opalinska J, Nooka AK. Safety and efficacy of belantamab mafodotin with pembrolizumab in patients with relapsed or refractory multiple myeloma. Cancer 2024; 130:2629-2641. [PMID: 38630908 DOI: 10.1002/cncr.35319] [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: 10/25/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Belantamab mafodotin (belamaf) has shown promising antimyeloma activity in relapsed or refractory multiple myeloma (RRMM) as a single agent. It was hypothesized that its multimodal activity may be enhanced by programmed cell death protein 1 pathway inhibition and activation of T cell-mediated antitumor responses. This study investigated the efficacy and safety of belamaf with pembrolizumab in patients with RRMM. METHODS DREAMM-4 (NCT03848845) was an open-label, single-arm, phase 1/2 study divided into dose-escalation (part 1) and dose-expansion (part 2) phases. Patients were ≥18 years old with ≥3 prior lines of therapy including a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 agent. Patients received belamaf (2.5 or 3.4 mg/kg, part 1; 2.5 mg/kg, part 2) and 200 mg pembrolizumab for ≤35 cycles. RESULTS Of 41 enrolled patients, 34 (n = 6 part 1, n = 28 part 2) who received 2.5 mg/kg belamaf plus pembrolizumab were included in this final analysis. Sixteen patients (47%) achieved an overall response. Minimal residual disease negativity was achieved in three of 10 patients who had very good partial response or better. Five of eight patients who had prior anti-B-cell maturation antigen therapy achieved partial response or better, including two who had B-cell maturation antigen-refractory disease. Common grade ≥3 adverse events were keratopathy (38%) and thrombocytopenia (29%). Despite belamaf-related ocular events, quality-of-life measures remained stable over time. No new safety signals were observed. CONCLUSIONS The results of DREAMM-4 demonstrated clinical activity and a favorable safety profile of belamaf plus pembrolizumab in patients with RRMM. This trial is registered at www. CLINICALTRIALS gov as NCT03848845.
Collapse
Affiliation(s)
- Attaya Suvannasankha
- Indiana University Simon Cancer Center and Roudebush VAMC, Indianapolis, Indiana, USA
| | - Nizar Bahlis
- Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Suzanne Trudel
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Katja Weisel
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Koenecke
- Hannover Medical School, Clinic for Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Albert Oriol
- Institut Català d'Oncologia and Institut Josep Carreras, Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Peter M Voorhees
- Levine Cancer Institute, Atrium Health/Wake Forest University School of Medicine, Charlotte, North Carolina, USA
| | | | | | - María-Victoria Mateos
- Instituto de Investigación Biomédica de Salamanca and Centro de Investigación del Cáncer, Hospital Universitario de Salamanca, Salamanca, Spain
| | | | - Shawn Hakim
- GlaxoSmithKline, Upper Providence, Pennsylvania, USA
| | | | | | | | | | | | - Ira Gupta
- GlaxoSmithKline, Philadelphia, Pennsylvania, USA
| | | | - Ajay K Nooka
- Winship Cancer Institute, Emory University Hospital, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Zhao J, Zheng M, Ma L, Guan T, Su L. From spear to trident: Upgrading arsenal of CAR-T cells in the treatment of multiple myeloma. Heliyon 2024; 10:e29997. [PMID: 38699030 PMCID: PMC11064441 DOI: 10.1016/j.heliyon.2024.e29997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Multiple myeloma (MM), marked by abnormal proliferation of plasma cells and production of monoclonal immunoglobulin heavy or light chains in the majority of patients, has traditionally been associated with poor survival, despite improvements achieved in median survival in all age groups since the introduction of novel agents. Survival has significantly improved with the development of new drugs and new treatment options, such as chimeric antigen receptor T-cell therapy (CAR-T), which have shown promise and given new hope in MM therapy. CARs are now classified as first-, second-, and third-generation CARs based on the number of monovalent to trivalent co-stimulatory molecules incorporated into their design. The scope of this review was relatively narrow because it was mainly about a comparison of the literature on the clinical application of CAR-T therapy in MM. Thus, our goal is to provide an overview of the new advances of CAR-T cells in the cure of MM, so in this review we looked at the progress of the clinical use of CAR-T cells in MM to try to provide a reference for their clinical use when managing MM.
Collapse
Affiliation(s)
| | | | - Li Ma
- Department of Hematology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, China
| | - Tao Guan
- Department of Hematology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, China
| | - Liping Su
- Department of Hematology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, China
| |
Collapse
|
4
|
Ailawadhi S, Shune L, Wong SW, Lin Y, Patel K, Jagannath S. Optimizing the CAR T-Cell Therapy Experience in Multiple Myeloma: Clinical Pearls From an Expert Roundtable. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:e217-e225. [PMID: 38369437 DOI: 10.1016/j.clml.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies offer substantial advancement in the treatment of multiple myeloma (MM). However, the CAR-T therapy process involves complex decision-making that is informed by many variables. This review aims to provide an overview of the patient selection and administration process for CAR-T therapy for MM from the perspective of experienced healthcare providers (HCPs), including considerations for each step in the CAR-T therapy process. Referring HCPs should initiate conversations with HCPs at CAR-T capable centers earlier in the treatment journey, even before patients are eligible for CAR-T therapy, particularly for patients from underserved populations and patients with high-risk disease, to ensure adequate time for logistical planning and patient education. Patient selection for CAR-T therapy may be guided by factors such as performance status, rate of disease progression, and logistical considerations. Some anticancer therapies may affect T-cell fitness and therefore impact CAR-T manufacturing and patient outcomes; however, additional research is needed to confirm this in MM. Bridging therapies should be tailored to the needs of the patient and ideally halted 1 week or longer before CAR-T infusion, contingent upon the agent(s) used. Lymphodepletion regimens may need to be modified for patients with renal insufficiency. Collaboration with HCPs at both the treating and referring centers is important to optimize coordinated care of patients. Collaboration with and guidance from experienced HCPs throughout patient selection, referral, and CAR-T administration is instrumental in optimizing patient outcomes as access to CAR-T therapies expands.
Collapse
Affiliation(s)
| | - Leyla Shune
- Division of Hematologic Malignancy and Cellular Therapeutics, University of Kansas Cancer Center, Westwood, Kansas
| | - Sandy W Wong
- Division of Hematology/Oncology, University of California San Francisco, San Francisco, CA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, Rochester, MN
| | - Krina Patel
- Department of Lymphoma - Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
5
|
Haran A, Vaxman I, Gatt ME, Lebel E. Immune Therapies in AL Amyloidosis-A Glimpse to the Future. Cancers (Basel) 2024; 16:1605. [PMID: 38672686 PMCID: PMC11048972 DOI: 10.3390/cancers16081605] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Light-chain (AL) amyloidosis is a rare plasma cell disorder characterized by the deposition of misfolded immunoglobulin light chains in target organs, leading to multi-organ dysfunction. Treatment approaches have historically mirrored but lagged behind those of multiple myeloma (MM). Recent advancements in MM immunotherapy are gradually being evaluated and adopted in AL amyloidosis. This review explores the current state of immunotherapeutic strategies in AL amyloidosis, including monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and chimeric antigen receptor T-cell therapy. We discuss the unique challenges and prospects of these therapies in AL amyloidosis, including the exposure of frail AL amyloidosis patients to immune-mediated toxicities such as cytokine release syndrome (CRS) and immune effector-cell-associated neurotoxicity syndrome (ICANS), as well as their efficacy in promoting rapid and deep hematologic responses. Furthermore, we highlight the need for international initiatives and compassionate programs to provide access to these promising therapies and address critical unmet needs in AL amyloidosis management. Finally, we discuss future directions, including optimizing treatment sequencing and mitigating toxicities, to improve outcomes for AL amyloidosis patients.
Collapse
Affiliation(s)
- Arnon Haran
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah Tikva 49100, Israel;
| | - Moshe E. Gatt
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| | - Eyal Lebel
- Department of Hematology, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; (A.H.); (M.E.G.)
| |
Collapse
|
6
|
St Martin Y, Franz JK, Agha ME, Lazarus HM. Failure of CAR-T cell therapy in relapsed and refractory large cell lymphoma and multiple myeloma: An urgent unmet need. Blood Rev 2023; 60:101095. [PMID: 37173224 DOI: 10.1016/j.blre.2023.101095] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Since its FDA approval, chimeric antigen receptor (CAR)-T cell therapy is changing the landscape of the treatment algorithm for relapsed and refractory large cell lymphoma and multiple myeloma. While initially hailed as a game changer and received widely with great enthusiasm, the reality of treatment failure soon became a major disappointment. This situation left patients and clinicians alike wondering about the next treatment options. CAR-T cell therapy failure for aggressive lymphoma or multiple myeloma creates a very poor prognosis and the treatment options are very limited. New emerging data, however, show promise for the use of approaches that include bispecific antibodies and other strategies to rescue affected patients. In this review, we summarize the current emerging data on the treatment options for patients whose disease has relapsed or remains refractory after CAR-T cell therapy failure, an area of great unmet need.
Collapse
Affiliation(s)
| | - Joseph K Franz
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America
| | - Mounzer E Agha
- University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, United States of America.
| | - Hillard M Lazarus
- Case Western Reserve University, Cleveland, OH, United States of America
| |
Collapse
|