1
|
Perra M, Castangia I, Aroffu M, Fulgheri F, Abi-Rached R, Manca ML, Cortés H, Del Prado-Audelo ML, Nomura-Contreras C, Romero-Montero A, Büsselberg D, Leyva-Gómez G, Sharifi-Rad J, Calina D. Maytansinoids in cancer therapy: advancements in antibody-drug conjugates and nanotechnology-enhanced drug delivery systems. Discov Oncol 2025; 16:73. [PMID: 39838217 PMCID: PMC11751265 DOI: 10.1007/s12672-025-01820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
Cancer remains the second leading cause of death globally, driving the need for innovative therapies. Among natural compounds, maytansinoids have shown significant promise, contributing to nearly 25% of recently approved anticancer drugs. Despite their potential, early clinical trials faced challenges due to severe side effects, prompting advancements in delivery systems such as antibody-maytansinoid conjugates (AMCs). This review highlights the anticancer activity of maytansinoids, with a focus on AMCs designed to target cancer cells specifically. Preclinical and clinical studies show that AMCs, including FDA-approved drugs like Kadcyla and Elahere, effectively inhibit tumor growth while reducing systemic toxicity. Key developments include improved synthesis methods, linker chemistry and payload design. Ongoing research aims to enhance the safety and efficacy of AMCs, integrate nanotechnology for drug delivery, and identify novel therapeutic targets. These advancements hold potential to transform maytansinoid-based cancer treatments in the future.
Collapse
Affiliation(s)
- Matteo Perra
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Ines Castangia
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Matteo Aroffu
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Federica Fulgheri
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Rita Abi-Rached
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy
| | - Maria Letizia Manca
- DISVA-Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. DeMonserrato-Sestu Km 0.700, 09042 CA, Monserrato, Italy.
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, Mexico
| | | | | | - Alejandra Romero-Montero
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
2
|
Pant A, Laliwala A, Holstein SA, Mohs AM. Recent advances in targeted drug delivery systems for multiple myeloma. J Control Release 2024; 376:215-230. [PMID: 39384153 PMCID: PMC11611669 DOI: 10.1016/j.jconrel.2024.10.003] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Despite significant therapeutic advances, multiple myeloma (MM) remains a challenging, incurable, hematological malignancy. The efficacy of traditional chemotherapy and currently available anti-MM agents is in part limited by their adverse effects, which restrict their therapeutic potential. Nanotherapeutics is an emerging field of cancer therapy that can overcome the biological and chemical barriers of existing anticancer drugs. This review presents an overview of recent advancements in nanoparticle- and immunotherapy-based drug delivery systems for MM treatment. It further delves into the targeting strategies, mechanism of controlled drug release, and challenges associated with the development of drug delivery systems for the treatment of MM.
Collapse
Affiliation(s)
- Ashruti Pant
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aayushi Laliwala
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Sarah A Holstein
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Internal Medicine, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE 68198, USA.
| |
Collapse
|
3
|
Anderson GSF, Chapman MA. T cell-redirecting therapies in hematological malignancies: Current developments and novel strategies for improved targeting. Mol Ther 2024; 32:2856-2891. [PMID: 39095991 PMCID: PMC11403239 DOI: 10.1016/j.ymthe.2024.07.028] [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: 03/25/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
T cell-redirecting therapies (TCRTs), such as chimeric antigen receptor (CAR) or T cell receptor (TCR) T cells and T cell engagers, have emerged as a highly effective treatment modality, particularly in the B and plasma cell-malignancy setting. However, many patients fail to achieve deep and durable responses; while the lack of truly unique tumor antigens, and concurrent on-target/off-tumor toxicities, have hindered the development of TCRTs for many other cancers. In this review, we discuss the recent developments in TCRT targets for hematological malignancies, as well as novel targeting strategies that aim to address these, and other, challenges.
Collapse
Affiliation(s)
| | - Michael A Chapman
- MRC Toxicology Unit, University of Cambridge, Cambridge CB2 1QR, UK; Department of Haematology, University of Cambridge, Cambridge CB2 0XY, UK; Addenbrooke's Hospital, Cambridge Universities Foundation Trust, Cambridge CB2 0QQ, UK.
| |
Collapse
|
4
|
Tan MSY, Chen Y, Smith EL. Beyond BCMA: newer immune targets in myeloma. Blood Adv 2024; 8:4433-4446. [PMID: 38865708 PMCID: PMC11375259 DOI: 10.1182/bloodadvances.2023010856] [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: 12/12/2023] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
ABSTRACT The identification and targeting of B-cell maturation antigen (BCMA) through immunotherapeutic strategies such as antibody-drug conjugates, chimeric antigen receptor T cells, and T-cell engagers have revolutionized the care of patients with multiple myeloma (MM). These treatment modalities have improved the survival outcomes of patients with relapsed and/or refractory MM compared with previously established strategies and are moving into earlier lines of therapy. Despite their efficacy, the majority of patients eventually relapse, necessitating additional therapeutic targets for salvage. G-protein-coupled receptor class 5 member D, Fc receptor-homolog 5, and SLAMF7 are some examples of novel targets in development. This expanding armamentarium of immunotherapeutic agents will be crucial to address the unmet need for relapses after BCMA-targeting therapies, particularly antigen-negative relapses. The utilization of sequential T-cell redirective therapies including agents targeting different tumor-associated antigens and combination therapies appears feasible, paving the way for effective chemotherapy-free regimes. Deliberate consideration of treatment timing, preserving T-cell health, overcoming antigenic loss, and comprehension of the complex tumor microenvironment would be key to maximizing therapeutic benefits and minimizing adverse effects. This review summarizes novel targets in development for myeloma beyond BCMA, presenting pivotal safety and efficacy data derived from clinical trials when available and the considerations vital for navigating this expanding landscape of immunotherapeutic options.
Collapse
Affiliation(s)
- Melinda S. Y. Tan
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Yunxin Chen
- Department of Haematology, Singapore General Hospital, Singapore
- Department of Haematology, National Cancer Centre Singapore, Singapore
| | - Eric L. Smith
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
5
|
Blake-Hedges J, Groff D, Foo W, Hanson J, Castillo E, Wen M, Cheung D, Masikat MR, Lu J, Park Y, Carlos NA, Usman H, Fong K, Yu A, Zhou S, Kwong J, Tran C, Li X, Yuan D, Hallam T, Yin G. Production of antibodies and antibody fragments containing non-natural amino acids in Escherichia coli. MAbs 2024; 16:2316872. [PMID: 38381460 PMCID: PMC10883104 DOI: 10.1080/19420862.2024.2316872] [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: 09/29/2023] [Accepted: 02/06/2024] [Indexed: 02/22/2024] Open
Abstract
Therapeutic bioconjugates are emerging as an essential tool to combat human disease. Site-specific conjugation technologies are widely recognized as the optimal approach for producing homogeneous drug products. Non-natural amino acid (nnAA) incorporation allows the introduction of bioconjugation handles at genetically defined locations. Escherichia coli (E. coli) is a facile host for therapeutic nnAA protein synthesis because it can stably replicate plasmids encoding genes for product and nnAA incorporation. Here, we demonstrate that by engineering E. coli to incorporate high levels of nnAAs, it is feasible to produce nnAA-containing antibody fragments and full-length immunoglobulin Gs (IgGs) in the cytoplasm of E. coli. Using high-density fermentation, it was possible to produce both of these types of molecules with site-specifically incorporated nnAAs at titers > 1 g/L. We anticipate this strategy will help simplify the production and manufacture of promising antibody therapeutics.
Collapse
Affiliation(s)
| | - Dan Groff
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Wilson Foo
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Jeffrey Hanson
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Elenor Castillo
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Miao Wen
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Diana Cheung
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Mary Rose Masikat
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Jian Lu
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Young Park
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Nina Abi Carlos
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Hans Usman
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Kevin Fong
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Abigail Yu
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Sihong Zhou
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Joyce Kwong
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Cuong Tran
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Xiaofan Li
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Dawei Yuan
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Trevor Hallam
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| | - Gang Yin
- Research and Process Development, Sutro Biopharma, Inc, South San Francisco, CA, USA
| |
Collapse
|
6
|
Vargas J, Pantouris G. Analysis of CD74 Occurrence in Oncogenic Fusion Proteins. Int J Mol Sci 2023; 24:15981. [PMID: 37958963 PMCID: PMC10650716 DOI: 10.3390/ijms242115981] [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: 09/17/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
CD74 is a type II cell surface receptor found to be highly expressed in several hematological and solid cancers, due to its ability to activate pathways associated with tumor cell survival and proliferation. Over the past 16 years, CD74 has emerged as a commonly detected fusion partner in multiple oncogenic fusion proteins. Studies have found CD74 fusion proteins in a range of cancers, including lung adenocarcinoma, inflammatory breast cancer, and pediatric acute lymphoblastic leukemia. To date, there are five known CD74 fusion proteins, CD74-ROS1, CD74-NTRK1, CD74-NRG1, CD74-NRG2α, and CD74-PDGFRB, with a total of 16 different variants, each with unique genetic signatures. Importantly, the occurrence of CD74 in the formation of fusion proteins has not been well explored despite the fact that ROS1 and NRG1 families utilize CD74 as the primary partner for the formation of oncogenic fusions. Fusion proteins known to be oncogenic drivers, including those of CD74, are typically detected and targeted after standard chemotherapeutic plans fail and the disease relapses. The analysis reported herein provides insights into the early intervention of CD74 fusions and highlights the need for improved routine assessment methods so that targeted therapies can be applied while they are most effective.
Collapse
Affiliation(s)
| | - Georgios Pantouris
- Department of Chemistry, University of the Pacific, Stockton, CA 95211, USA;
| |
Collapse
|
7
|
Riccardi F, Dal Bo M, Macor P, Toffoli G. A comprehensive overview on antibody-drug conjugates: from the conceptualization to cancer therapy. Front Pharmacol 2023; 14:1274088. [PMID: 37790810 PMCID: PMC10544916 DOI: 10.3389/fphar.2023.1274088] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 10/05/2023] Open
Abstract
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-cancer compounds that are widely used in the treatment of hematologic malignancies and solid tumors. Unlike conventional chemotherapeutic drug-based therapies, that are mainly associated with modest specificity and therapeutic benefit, the three key components that form an ADC (a monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety) achieve remarkable improvement in terms of targeted killing of cancer cells and, while sparing healthy tissues, a reduction in systemic side effects caused by off-tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been approved to date by the market approval by the Food and Drug Administration (FDA), the European Medicines Agency (EMA) and/or other international governmental agencies for use in clinical oncology, and hundreds are undergoing evaluation in the preclinical and clinical phases. Here, our aim is to provide a comprehensive overview of the key features revolving around ADC therapeutic strategy including their structural and targeting properties, mechanism of action, the role of the tumor microenvironment and review the approved ADCs in clinical oncology, providing discussion regarding their toxicity profile, clinical manifestations and use in novel combination therapies. Finally, we briefly review ADCs in other pathological contexts and provide key information regarding ADC manufacturing and analytical characterization.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano, Italy
| |
Collapse
|
8
|
Ray U, Orlowski RZ. Antibody-Drug Conjugates for Multiple Myeloma: Just the Beginning, or the Beginning of the End? Pharmaceuticals (Basel) 2023; 16:ph16040590. [PMID: 37111346 PMCID: PMC10145905 DOI: 10.3390/ph16040590] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Multiple myeloma is a malignancy of immunoglobulin-secreting plasma cells that is now often treated in the newly diagnosed and relapsed and/or refractory settings with monoclonal antibodies targeting lineage-specific markers used either alone or in rationally designed combination regimens. Among these are the anti-CD38 antibodies daratumumab and isatuximab, and the anti-Signaling lymphocytic activation molecule family member 7 antibody elotuzumab, all of which are used in their unconjugated formats. Single-chain variable fragments from antibodies also form a key element of the chimeric antigen receptors (CARs) in the B-cell maturation antigen (BCMA)-targeted CAR T-cell products idecabtagene vicleucel and ciltacabtagene autoleucel, which are approved in the advanced setting. Most recently, the bispecific anti-BCMA and T-cell-engaging antibody teclistamab has become available, again for patients with relapsed/refractory disease. Another format into which antibodies can be converted to exert anti-tumor efficacy is as antibody-drug conjugates (ADCs), and belantamab mafodotin, which also targets BCMA, represented the first such agent that gained a foothold in myeloma. Negative results from a recent Phase III study have prompted the initiation of a process for withdrawal of its marketing authorization. However, belantamab remains a drug with some promise, and many other ADCs targeting either BCMA or other plasma cell surface markers are in development and showing potential. This contribution will provide an overview of some of the current data supporting the possibility that ADCs will remain a part of our chemotherapeutic armamentarium against myeloma moving forward, and also highlight areas for future development.
Collapse
Affiliation(s)
- Upasana Ray
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 429, Houston, TX 77030-4009, USA
| |
Collapse
|
9
|
Morè S, Corvatta L, Manieri VM, Olivieri A, Offidani M. Current Main Topics in Multiple Myeloma. Cancers (Basel) 2023; 15:2203. [PMID: 37190132 PMCID: PMC10136770 DOI: 10.3390/cancers15082203] [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: 02/14/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Multiple Myeloma (MM) remains a difficult to treat disease mainly due to its biological heterogeneity, of which we are more and more knowledgeable thanks to the development of increasingly sensitive molecular methods that allow us to build better prognostication models. The biological diversity translates into a wide range of clinical outcomes from long-lasting remission in some patients to very early relapse in others. In NDMM transplant eligible (TE) patients, the incorporation of mAb as daratumumab in the induction regimens, followed by autologous stem cell transplantation (ASCT) and consolidation/maintenance therapy, has led to a significant improvement of PFS and OS.; however, this outcome remains poor in ultra-high risk MM or in those who did not achieve a minimal residual disease (MRD) negativity. Several trials are exploring cytogenetic risk-adapted and MRD-driven therapies in these patients. Similarly, quadruplets-containing daratumumab, particularly when administered as continuous therapies, have improved outcome of patients not eligible for autologous transplant (NTE). Patients who become refractory to conventional therapies have noticeably poor outcomes, making their treatment a difficult challenge in need of novel strategies. In this review, we will focus on the main points regarding risk stratification, treatment and monitoring of MM, highlighting the most recent evidence that could modify the management of this still incurable disease.
Collapse
Affiliation(s)
- Sonia Morè
- Clinica di Ematologia Azienda Ospedaliero, Universitaria delle Marche, 60126 Ancona, Italy
| | - Laura Corvatta
- Unità Operativa Complessa di Medicina, Ospedale Profili, 60044 Fabriano, Italy
| | | | - Attilio Olivieri
- 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
|
10
|
Li X, Abrahams C, Yu A, Embry M, Henningsen R, DeAlmeida V, Matheny S, Kline T, Yam A, Stafford R, Hallam T, Lupher M, Molina A. Targeting CD74 in B-cell non-Hodgkin lymphoma with the antibody-drug conjugate STRO-001. Oncotarget 2023; 14:1-13. [PMID: 36634212 PMCID: PMC9836384 DOI: 10.18632/oncotarget.28341] [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] [Indexed: 01/14/2023] Open
Abstract
Overexpression of CD74, a type II transmembrane glycoprotein involved in MHC class II antigen presentation, has been reported in many B-cell non-Hodgkin lymphomas (NHLs) and in multiple myeloma (MM). STRO-001 is a site-specific, predominantly single-species antibody-drug conjugate (ADC) that targets CD74 and has demonstrated efficacy in xenograft models of MM and tolerability in non-human primates. Here we report results of preclinical studies designed to elucidate the potential role of STRO-001 in B-cell NHL. STRO-001 displayed nanomolar and sub-nanomolar cytotoxicity in 88% (15/17) of cancer cell lines tested. STRO-001 showed potent cytotoxicity on proliferating B cells while limited cytotoxicity was observed on naïve human B cells. A linear dose-response relationship was demonstrated in vivo for DLBCL models SU-DHL-6 and U2932. Tumor regression was induced at doses less than 5 mg/kg, while maximal activity with complete cures were observed starting at 10 mg/kg. In MCL Mino and Jeko-1 xenografts, STRO-001 starting at 3 mg/kg significantly prolonged survival or induced tumor regression, respectively, leading to tumor eradication in both models. In summary, high CD74 expression levels in tumors, nanomolar cellular potency, and significant anti-tumor in DLBCL and MCL xenograft models support the ongoing clinical study of STRO-001 in patients with B-cell NHL.
Collapse
Affiliation(s)
- Xiaofan Li
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA,Correspondence to:Xiaofan Li, email:
| | - Cristina Abrahams
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Abigail Yu
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Millicent Embry
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Robert Henningsen
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Venita DeAlmeida
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Shannon Matheny
- 2Clinical Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Toni Kline
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Alice Yam
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Ryan Stafford
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Trevor Hallam
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Mark Lupher
- 1Research Development, Sutro Biopharma, South San Francisco, CA 94080, USA
| | - Arturo Molina
- 2Clinical Development, Sutro Biopharma, South San Francisco, CA 94080, USA,Arturo Molina, email:
| |
Collapse
|
11
|
Tannoury M, Garnier D, Susin SA, Bauvois B. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What's Coming Next? Cancers (Basel) 2022; 14:6026. [PMID: 36551511 PMCID: PMC9775488 DOI: 10.3390/cancers14246026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today's commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton's tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody-drug conjugates, antibody-radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.
Collapse
Affiliation(s)
| | | | | | - Brigitte Bauvois
- Centre de Recherche des Cordeliers, Sorbonne Université, Université Paris Cité, Inserm, Cell Death and Drug Resistance in Lymphoproliferative Disorders Team, F-75006 Paris, France
| |
Collapse
|
12
|
Antibody Drug Conjugates in Multiple Myeloma. Cancer J 2022; 28:488-495. [DOI: 10.1097/ppo.0000000000000628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Bautista L, Pill-Pepe L, Kapoor N, Snyder S, Chu E, Agarwal P, Sardar M, Arulkumar S, Berges A, Iverson M, Behrens C, Marcq O, Fairman J. Addition of Lauryldimethylamine N-Oxide (LDAO) to a Copper-Free Click Chemistry Reaction Improves the Conjugation Efficiency of a Cell-Free Generated CRM197 Variant to Clinically Important Streptococcus pneumoniae Serotypes. ACS OMEGA 2022; 7:34921-34928. [PMID: 36211053 PMCID: PMC9535640 DOI: 10.1021/acsomega.2c03481] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/07/2022] [Indexed: 05/22/2023]
Abstract
Strain-promoted azide-alkyne cycloaddition (SPAAC) reactions like click chemistry have the potential to be highly scalable, robust, and cost-effective methods for generating small- and large-molecule conjugates for a variety of applications. However, despite method improvements, the rates of copper-based click chemistry reactions continue to be much faster than the rates of copper-free click chemistry reactions, which makes broader deployment of click chemistry challenging from a safety and compatibility standpoint. In this study, we used a zwitterionic detergent, namely, lauryldimethylamine N-oxide (LDAO), in a copper-free click chemistry reaction to investigate its impact on the generation of conjugate vaccines (CVs). For this, we utilized an Xpress cell-free protein synthesis (CFPS) platform to generate a proprietary variant of CRM197 (eCRM) containing non-native amino acids (nnAA) with azide-containing side chains as a carrier protein for conjugation to several clinically relevant dibenzocyclooctyne (DBCO)-derivatized S. pneumoniae serotypes (types 3, 5, 18C, and 19A). For conjugation, we performed copper-free click chemistry in the presence and absence of LDAO. Our results show that the addition of LDAO significantly enhanced the reaction kinetics to generate larger conjugates, which were similarly immunogenic and equally stable to conjugates generated without LDAO. Most importantly, the addition of LDAO substantially improved the efficiency of the conjugation process. Thus, our results for the first time show that the addition of a zwitterionic surfactant to a copper-free click chemistry reaction can significantly accelerate the reaction kinetics along with improving the efficiency of the conjugation process.
Collapse
|
14
|
Dima D, Jiang D, Singh DJ, Hasipek M, Shah HS, Ullah F, Khouri J, Maciejewski JP, Jha BK. Multiple Myeloma Therapy: Emerging Trends and Challenges. Cancers (Basel) 2022; 14:cancers14174082. [PMID: 36077618 PMCID: PMC9454959 DOI: 10.3390/cancers14174082] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a complex hematologic malignancy characterized by the uncontrolled proliferation of clonal plasma cells in the bone marrow that secrete large amounts of immunoglobulins and other non-functional proteins. Despite decades of progress and several landmark therapeutic advancements, MM remains incurable in most cases. Standard of care frontline therapies have limited durable efficacy, with the majority of patients eventually relapsing, either early or later. Induced drug resistance via up-modulations of signaling cascades that circumvent the effect of drugs and the emergence of genetically heterogeneous sub-clones are the major causes of the relapsed-refractory state of MM. Cytopenias from cumulative treatment toxicity and disease refractoriness limit therapeutic options, hence creating an urgent need for innovative approaches effective against highly heterogeneous myeloma cell populations. Here, we present a comprehensive overview of the current and future treatment paradigm of MM, and highlight the gaps in therapeutic translations of recent advances in targeted therapy and immunotherapy. We also discuss the therapeutic potential of emerging preclinical research in multiple myeloma.
Collapse
Affiliation(s)
- Danai Dima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dongxu Jiang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Divya Jyoti Singh
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Metis Hasipek
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Haikoo S. Shah
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fauzia Ullah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Babal K. Jha
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
- Correspondence:
| |
Collapse
|
15
|
Zawada JF, Burgenson D, Yin G, Hallam TJ, Swartz JR, Kiss RD. Cell-free technologies for biopharmaceutical research and production. Curr Opin Biotechnol 2022; 76:102719. [DOI: 10.1016/j.copbio.2022.102719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/15/2022] [Accepted: 03/02/2022] [Indexed: 11/03/2022]
|
16
|
Peng F, Yan S, Liu H, Liu Z, Jiang F, Cao P, Fu R. Roles of LINC01473 and CD74 in osteoblasts in multiple myeloma bone disease. J Investig Med 2022; 70:1301-1307. [PMID: 35145037 PMCID: PMC9240337 DOI: 10.1136/jim-2021-002192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2022] [Indexed: 11/22/2022]
Abstract
The suppression of osteoblast (OB) activity is partially responsible for multiple myeloma (MM) bone disease. Long non-coding RNAs (lncRNAs) play a vital role in bone formation and resorption. However, their functions in OBs from patients with MM have rarely been reported. Through high-throughput sequencing of OBs from patients with MM and healthy controls, we identified several lncRNAs and messenger RNAs (mRNAs) with different expression profile and validated them using quantitative real-time PCR. In total, 22 upregulated and 21 downregulated lncRNAs were found in OBs from patients with MM. Moreover, 18 upregulated protein-coding mRNAs were identified. The expression levels of LINC01473 and its associated co-expression mRNA, CD74, were higher in patients with MM than in healthy controls (p=0.047 and p=0.016, respectively). LINC01473 expression demonstrated a negative correlation with serum interleukin-2 and tumor necrosis factor α levels, whereas the expression of mRNA CD74 was positively associated with serum lactic dehydrogenase in patients with MM. Aberrant expression of lncRNAs and mRNAs was observed in OBs from patients with MM. This study identifies new promising targets for further research on imbalanced bone formation and resorption and MM immune escape.
Collapse
Affiliation(s)
- Fengping Peng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Siyang Yan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengjuan Jiang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Panpan Cao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
17
|
Hartley-Brown M, Richardson P. Antibody-drug conjugate therapies in multiple myeloma—what’s next on the horizon? EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:1-10. [PMID: 36046359 PMCID: PMC9400726 DOI: 10.37349/etat.2022.00067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Targeted immunotherapy has arisen over the past decade to the forefront of cancer care. Notably, targeted therapies such as antibody-drug conjugates (ADCs) are becoming more recognized for a novel approach in cancer treatment. The mechanism of action of ADCs incorporates a monoclonal antibody portion directed against the tumor cell antigen and attached to the tumoricidal portion via chemical linkage. The binding of the monoclonal antibody portion allows for tumor cell internalization of the ADC and precise release of the toxic payload within the cancer cell. Multiple myeloma (MM) is an incurable cancer for which belantamab mafodotin was the first-in-class ADC to achieve United States Food and Drug Administration (FDA) approval for treatment of this disease. Clinical trials are currently evaluating other ADCs in the treatment of MM. In this review, a look at the current ADCs being tested in MM clinical trials with a focus on those that are more promising and a potential next-in-line for FDA approval for treatment of MM is discussed.
Collapse
Affiliation(s)
- Monique Hartley-Brown
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Paul Richardson
- Department of Medicine, Division of Medical Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
18
|
Thooft K, Van Breedam W, Santens F, Wyseure E, Vanmarcke S, Devos S, Callewaert N, Madder A. GlyConnect-Ugi: site-selective, multi-component glycoprotein conjugations through GlycoDelete expressed glycans. Org Biomol Chem 2022; 20:464-471. [PMID: 34913461 DOI: 10.1039/d1ob02299g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recently, the GlyConnect-oxime (GC) protein conjugation strategy was developed to provide a site-selective glycan-based conjugation strategy as an extension to the in-house developed GlycoDelete (GD) technology. GD gives access to glycoproteins with single GlcNAc, LacNAc, or LacNAc-Sia type glycans on their N-glycosylation sites. We have previously shown that these glycans provide a unique handle for site-selective conjugation as they provide a short, homogeneous and hydrophilic link to the protein backbone. GC focused on the use of chemical and chemo-enzymatic pathways for conjugation of a single molecule of interest via oxime formation or reductive amination. In the current work, we explore multicomponent reactions (MCR), namely Ugi and Passerini reactions, for GlycoDelete glycan directed, site-specific protein conjugation (MC-GC). The use of the Ugi and Passerini multicomponent reactions holds the potential of introducing multiple groups of interest in a single reaction step while creating a hydrophilic peptide-like linker.
Collapse
Affiliation(s)
- Karel Thooft
- Organic and Biomimetic Chemistry Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Belgium. .,Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Wander Van Breedam
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Francis Santens
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Elise Wyseure
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Sandrine Vanmarcke
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Simon Devos
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Nico Callewaert
- Medical Biotechnology Centre, VIB, Ghent, Belgium.,Center for Medical Biotechnology, VIB, Department of Biochemistry and Microbiology, UGent, Technologiepark 75, B-9052 Zwijnaarde-Gent
| | - Annemieke Madder
- Organic and Biomimetic Chemistry Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Belgium.
| |
Collapse
|
19
|
Ceci C, Lacal PM, Graziani G. Antibody-drug conjugates: Resurgent anticancer agents with multi-targeted therapeutic potential. Pharmacol Ther 2022; 236:108106. [PMID: 34990642 DOI: 10.1016/j.pharmthera.2021.108106] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Antibody-drug conjugates (ADCs) constitute a relatively new group of anticancer agents, whose first appearance took place about two decades ago, but a renewed interest occurred in recent years, following the success of anti-cancer immunotherapy with monoclonal antibodies. Indeed, an ADC combines the selectivity of a monoclonal antibody with the cell killing properties of a chemotherapeutic agent (payload), joined together through an appropriate linker. The antibody moiety targets a specific cell surface antigen expressed by tumor cells and/or cells of the tumor microenvironment and acts as a carrier that delivers the cytotoxic payload within the tumor mass. Despite advantages in terms of selectivity and potency, the development of ADCs is not devoid of challenges, due to: i) low tumor selectivity when the target antigens are not exclusively expressed by cancer cells; ii) premature release of the cytotoxic drug into the bloodstream as a consequence of linker instability; iii) development of tumor resistance mechanisms to the payload. All these factors may result in lack of efficacy and/or in no safety improvement compared to unconjugated cytotoxic agents. Nevertheless, the development of antibodies engineered to remain inert until activated in the tumor (e.g., antibodies activated proteolytically after internalization or by the acidic conditions of the tumor microenvironment) together with the discovery of innovative targets and cytotoxic or immunomodulatory payloads, have allowed the design of next-generation ADCs that are expected to possess improved therapeutic properties. This review provides an overview of approved ADCs, with related advantages and limitations, and of novel targets exploited by ADCs that are presently under clinical investigation.
Collapse
Affiliation(s)
- Claudia Ceci
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | - Grazia Graziani
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy.
| |
Collapse
|
20
|
Rashid MH. Full-length recombinant antibodies from Escherichia coli: production, characterization, effector function (Fc) engineering, and clinical evaluation. MAbs 2022; 14:2111748. [PMID: 36018829 PMCID: PMC9423848 DOI: 10.1080/19420862.2022.2111748] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Although several antibody fragments and antibody fragment-fusion proteins produced in Escherichia coli (E. coli) are approved as therapeutics for various human diseases, a full-length monoclonal or a bispecific antibody produced in E. coli has not yet been approved. The past decade witnessed substantial progress in expression of full-length antibodies in the E. coli cytoplasm and periplasm, as well as in cell-free expression systems. The equivalency of E. coli-produced aglycosylated antibodies and their mammalian cell-produced counterparts, with respect to biochemical and biophysical properties, including antigen binding, in vitro and in vivo serum stability, pharmacokinetics, and in vivo serum half-life, has been demonstrated. Extensive engineering of the Fc domain of aglycosylated antibodies enables recruitment of various effector functions, despite the lack of N-linked glycans. This review summarizes recent research, preclinical advancements, and clinical development of E. coli-produced aglycosylated therapeutic antibodies as monoclonal, bispecific, and antibody-drug conjugates for use in autoimmune, oncology, and immuno-oncology areas.Abbreviations: ADA Anti-drug antibody; ADCC Antibody-dependent cellular cytotoxicity; ADCP Antibody-dependent cellular phagocytosis; ADC Antibody-drug conjugate; aFc Aglycosylated Fc; AMD Age-related macular degeneration aTTP Acquired thrombotic thrombocytopenic purpura; BCMA B-cell maturation antigen; BLA Biologics license application; BsAb Bispecific antibody; C1q Complement protein C1q; CDC Complement-dependent cytotoxicity; CDCC Complement-dependent cellular cytotoxicity; CDCP Complement-dependent cellular phagocytosis; CEX Cation exchange chromatography; CFPS Cell-free protein expression; CHO Chinese Hamster Ovary; CH1-3 Constant heavy chain 1-3; CL Constant light chain; DLBCL Diffuse large B-cell lymphoma; DAR Drug antibody ratio; DC Dendritic cell; dsFv Disulfide-stabilized Fv; EU European Union; EGFR Epidermal growth factor receptor; E. coli Escherichia coli; EpCAM Epithelial cell adhesion molecule; Fab Fragment antigen binding; FACS Fluorescence activated cell sorting; Fc Fragment crystallizable; FcRn Neonatal Fc receptor; FcɣRs Fc gamma receptors; FDA Food and Drug Administration; FL-IgG Full-length immunoglobulin; Fv Fragment variable; FolRαa Folate receptor alpha; gFc Glycosylated Fc; GM-CSF Granulocyte macrophage-colony stimulating factor; GPx7 Human peroxidase 7; HCL Hairy cell leukemia; HIV Human immunodeficiency virusl; HER2 Human epidermal growth factor receptor 2; HGF Hepatocyte growth factor; HIC Hydrophobic interaction chromatography; HLA Human leukocyte antigen; IBs Inclusion bodies; IgG1-4 Immunoglobulin 1-4; IP Intraperitoneal; ITC Isothermal titration calorimetry; ITP Immune thrombocytopenia; IV Intravenous; kDa Kilodalton; KiH Knob-into-Hole; mAb Monoclonal antibody; MAC Membrane-attack complex; mCRC Metastatic colorectal cancer; MM Multipl myeloma; MOA Mechanism of action; MS Mass spectrometry; MUC1 Mucin 1; MG Myasthenia gravis; NB Nanobody; NK Natural killer; nsAA Nonstandard amino acid; NSCLC Non-small cell lung cancer; P. aeruginosa Pseudomonas aeruginosa; PD-1 Programmed cell death 1; PD-L1 Programmed cell death-ligand 1; PDI Protein disulfide isomerase; PECS Periplasmic expression cytometric screening; PK Pharmacokinetics; P. pastoris Pichia pastoris; PTM Post-translational modification; Rg Radius of gyration; RA Rheumatoid arthritis; RT-PCR Reverse transcription polymerase chain reaction; SAXS Small angle X-ray scattering; scF Single chain Fv; SCLC Small cell lung cancer; SDS-PAGE Sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SEC Size exclusion chromatography; SEED Strand-exchange engineered domain; sRNA Small regulatory RNA; SRP Signal recognition particle; T1/2 Half-life; Tagg Aggregation temperature; TCR T cell receptor; TDB T cell-dependent bispecific; TF Tissue factor; TIR Translation initiation region; Tm Melting temperature; TNBC Triple-negative breast cancer; TNF Tumor necrosis factor; TPO Thrombopoietin; VEGF Vascular endothelial growth factor; vH Variable heavy chain; vL Variable light chain; vWF von Willebrand factor; WT Wild type.
Collapse
|
21
|
Efficient production of immunologically active Shigella invasion plasmid antigens IpaB and IpaH using a cell-free expression system. Appl Microbiol Biotechnol 2021; 106:401-414. [PMID: 34932164 PMCID: PMC8688910 DOI: 10.1007/s00253-021-11701-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022]
Abstract
Abstract Shigella spp. invade the colonic epithelium and cause bacillary dysentery in humans. Individuals living in areas that lack access to clean water and sanitation are the most affected. Even though infection can be treated with antibiotics, Shigella antimicrobial drug resistance complicates clinical management. Despite decades of effort, there are no licensed vaccines to prevent shigellosis. The highly conserved invasion plasmid antigens (Ipa), which are components of the Shigella type III secretion system, participate in bacterial epithelial cell invasion and have been pursued as vaccine targets. However, expression and purification of these proteins in conventional cell-based systems have been challenging due to solubility issues and extremely low recovery yields. These difficulties have impeded manufacturing and clinical advancement. In this study, we describe a new method to express Ipa proteins using the Xpress+TM cell-free protein synthesis (CFPS) platform. Both IpaB and the C-terminal domain of IpaH1.4 (IpaH-CTD) were efficiently produced with this technology at yields > 200 mg/L. Furthermore, the expression was linearly scaled in a bioreactor under controlled conditions, and proteins were successfully purified using multimode column chromatography to > 95% purity as determined by SDS-PAGE. Biophysical characterization of the cell-free synthetized IpaB and IpaH-CTD using SEC-MALS analysis showed well-defined oligomeric states of the proteins in solution. Functional analysis revealed similar immunoreactivity as compared to antigens purified from E. coli. These results demonstrate the efficiency of CFPS for Shigella protein production; the practicality and scalability of this method will facilitate production of antigens for Shigella vaccine development and immunological analysis. Key points • First report of Shigella IpaB and IpaH produced at high purity and yield using CFPS • CFPS-IpaB and IpaH perform similarly to E. coli–produced proteins in immunoassays • CFPS-IpaB and IpaH react with Shigella-specific human antibodies and are immunogenic in mice. Graphical abstract ![]()
Collapse
|
22
|
Su Z, Xiao D, Xie F, Liu L, Wang Y, Fan S, Zhou X, Li S. Antibody-drug conjugates: Recent advances in linker chemistry. Acta Pharm Sin B 2021; 11:3889-3907. [PMID: 35024314 PMCID: PMC8727783 DOI: 10.1016/j.apsb.2021.03.042] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/17/2021] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Antibody–drug conjugates (ADCs) are gradually revolutionizing clinical cancer therapy. The antibody–drug conjugate linker molecule determines both the efficacy and the adverse effects, and so has a major influence on the fate of ADCs. An ideal linker should be stable in the circulatory system and release the cytotoxic payload specifically in the tumor. However, existing linkers often release payloads nonspecifically and inevitably lead to off-target toxicity. This defect is becoming an increasingly important factor that restricts the development of ADCs. The pursuit of ADCs with optimal therapeutic windows has resulted in remarkable progress in the discovery and development of novel linkers. The present review summarizes the advance of the chemical trigger, linker‒antibody attachment and linker‒payload attachment over the last 5 years, and describes the ADMET properties of ADCs. This work also helps clarify future developmental directions for the linkers.
Collapse
Affiliation(s)
- Zheng Su
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fei Xie
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lianqi Liu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yanming Wang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shiyong Fan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Xinbo Zhou
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Corresponding author. Tel: +86 10 66930603 (Shiyong Fan), +86 10 66930673 (Xinbo Zhou).
| | - Song Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
23
|
Groff D, Carlos NA, Chen R, Hanson JA, Liang S, Armstrong S, Li X, Zhou S, Steiner A, Hallam TJ, Yin G. Development of an E. coli strain for cell-free ADC manufacturing. Biotechnol Bioeng 2021; 119:162-175. [PMID: 34655229 PMCID: PMC9297987 DOI: 10.1002/bit.27961] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022]
Abstract
Recent advances in cell‐free protein synthesis have enabled the folding and assembly of full‐length antibodies at high titers with extracts from prokaryotic cells. Coupled with the facile engineering of the Escherichia coli translation machinery, E. coli based in vitro protein synthesis reactions have emerged as a leading source of IgG molecules with nonnatural amino acids incorporated at specific locations for producing homogeneous antibody–drug conjugates (ADCs). While this has been demonstrated with extract produced in batch fermentation mode, continuous extract fermentation would facilitate supplying material for large‐scale manufacturing of protein therapeutics. To accomplish this, the IgG‐folding chaperones DsbC and FkpA, and orthogonal tRNA for nonnatural amino acid production were integrated onto the chromosome with high strength constitutive promoters. This enabled co‐expression of all three factors at a consistently high level in the extract strain for the duration of a 5‐day continuous fermentation. Cell‐free protein synthesis reactions with extract produced from cells grown continuously yielded titers of IgG containing nonnatural amino acids above those from extract produced in batch fermentations. In addition, the quality of the synthesized IgGs and the potency of ADC produced with continuously fermented extract were indistinguishable from those produced with the batch extract. These experiments demonstrate that continuous fermentation of E. coli to produce extract for cell‐free protein synthesis is feasible and helps unlock the potential for cell‐free protein synthesis as a platform for biopharmaceutical production.
Collapse
Affiliation(s)
- Dan Groff
- Sutro Biopharma, Inc., San Francisco, California, USA
| | - Nina A Carlos
- Sutro Biopharma, Inc., San Francisco, California, USA
| | - Rishard Chen
- Sutro Biopharma, Inc., San Francisco, California, USA
| | | | | | | | - Xiaofan Li
- Sutro Biopharma, Inc., San Francisco, California, USA
| | - Sihong Zhou
- Sutro Biopharma, Inc., San Francisco, California, USA
| | - Alex Steiner
- Sutro Biopharma, Inc., San Francisco, California, USA
| | | | - Gang Yin
- Sutro Biopharma, Inc., San Francisco, California, USA
| |
Collapse
|
24
|
Jadoon Y, Siddiqui MA. Immunotherapy in multiple myeloma. Cancer Treat Res Commun 2021; 29:100468. [PMID: 34653747 DOI: 10.1016/j.ctarc.2021.100468] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/19/2021] [Accepted: 09/25/2021] [Indexed: 02/03/2023]
Abstract
Despite available therapies, Multiple Myeloma (MM) remains an incurable hematologic malignancy. Over the past three decades, there have been tremendous developments in therapeutic options for MM. In regards to immunotherapy, Daratumumab was the first monoclonal antibody to receive FDA approval for multiple myeloma. Since then, other monoclonal antibodies such as elotuzumab and isatuximab have received FDA approval. Many clinical trials are underway investigating the efficacy of newer immunotherapies. This review summarizes recently presented and/or published data regarding this growing field, specifically regarding monoclonal antibodies, antibody-drug conjugates, bispecific antibodies, and trispecific antibodies.
Collapse
Affiliation(s)
- Yamna Jadoon
- Division of Hematology, Mayo Clinic, Rochester MN, United States of America
| | - Mustaqeem A Siddiqui
- Division of Hematology, Mayo Clinic, Rochester MN, United States of America; Division of Oncology and Hematology, Sheikh Shakhbout Medical City, Abu Dhabi, UAE.
| |
Collapse
|
25
|
Wurster KD, Costanza M, Kreher S, Glaser S, Lamprecht B, Schleussner N, Anagnostopoulos I, Hummel M, Jöhrens K, Stein H, Molina A, Diepstra A, Gillissen B, Köchert K, Siebert R, Merkel O, Kenner L, Janz M, Mathas S. Aberrant Expression of and Cell Death Induction by Engagement of the MHC-II Chaperone CD74 in Anaplastic Large Cell Lymphoma (ALCL). Cancers (Basel) 2021; 13:cancers13195012. [PMID: 34638496 PMCID: PMC8507667 DOI: 10.3390/cancers13195012] [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: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Anaplastic large cell lymphoma (ALCL) is a lymphoid malignancy considered to be derived from T cells. Currently, two types of systemic ALCL are distinguished: anaplastic lymphoma kinase (ALK)-positive and ALK-negative ALCL. Although ALK+ and ALK− ALCL differ at the genomic and molecular levels, various key biological and molecular features are highly similar between both entities. We have developed the concept that both ALCL entities share a common principle of pathogenesis. In support of this concept, we here describe a common deregulation of CD74, which is usually not expressed in T cells, in ALCL. Ligation of CD74 induces cell death of ALCL cells in various conditions, and an anti-CD74-directed antibody-drug conjugate efficiently kills ALCL cell lines. Furthermore, we reveal expression of the proto-oncogene and known CD74 interaction partner MET in a fraction of ALCL cases. These data give insights into ALCL pathogenesis and might help to develop new treatment strategies for ALCL. Abstract In 50–60% of cases, systemic anaplastic large cell lymphoma (ALCL) is characterized by the t(2;5)(p23;q35) or one of its variants, considered to be causative for anaplastic lymphoma kinase (ALK)-positive (ALK+) ALCL. Key pathogenic events in ALK-negative (ALK−) ALCL are less well defined. We have previously shown that deregulation of oncogenic genes surrounding the chromosomal breakpoints on 2p and 5q is a unifying feature of both ALK+ and ALK− ALCL and predisposes for occurrence of t(2;5). Here, we report that the invariant chain of the MHC-II complex CD74 or li, which is encoded on 5q32, can act as signaling molecule, and whose expression in lymphoid cells is usually restricted to B cells, is aberrantly expressed in T cell-derived ALCL. Accordingly, ALCL shows an altered DNA methylation pattern of the CD74 locus compared to benign T cells. Functionally, CD74 ligation induces cell death of ALCL cells. Furthermore, CD74 engagement enhances the cytotoxic effects of conventional chemotherapeutics in ALCL cell lines, as well as the action of the ALK-inhibitor crizotinib in ALK+ ALCL or of CD95 death-receptor signaling in ALK− ALCL. Additionally, a subset of ALCL cases expresses the proto-oncogene MET, which can form signaling complexes together with CD74. Finally, we demonstrate that the CD74-targeting antibody-drug conjugate STRO-001 efficiently and specifically kills CD74-positive ALCL cell lines in vitro. Taken together, these findings enabled us to demonstrate aberrant CD74-expression in ALCL cells, which might serve as tool for the development of new treatment strategies for this lymphoma entity.
Collapse
Affiliation(s)
- Kathrin D. Wurster
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Mariantonia Costanza
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Stephan Kreher
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University, Ulm University Medical Center, 89081 Ulm, Germany; (S.G.); (R.S.)
| | - Björn Lamprecht
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Nikolai Schleussner
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Ioannis Anagnostopoulos
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | - Michael Hummel
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | - Korinna Jöhrens
- Institute of Pathology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany; (I.A.); (K.J.)
| | | | - Arturo Molina
- Sutro Biopharma, South San Francisco, CA 94080, USA;
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, 9700 RB Groningen, The Netherlands;
| | - Bernd Gillissen
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Karl Köchert
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University, Ulm University Medical Center, 89081 Ulm, Germany; (S.G.); (R.S.)
| | - Olaf Merkel
- Unit of Experimental and Laboratory Animal Pathology, Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (O.M.); (L.K.)
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
| | - Lukas Kenner
- Unit of Experimental and Laboratory Animal Pathology, Department of Pathology, Medical University of Vienna, 1090 Vienna, Austria; (O.M.); (L.K.)
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
| | - Martin Janz
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
| | - Stephan Mathas
- Max-Delbrück-Center (MDC) for Molecular Medicine, 13125 Berlin, Germany; (M.C.); (N.S.); (M.J.)
- Department of Hematology, Oncology and Cancer Immunology, Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12200 Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation between the Charité and the MDC, 13125 Berlin, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- European Research Initiative on ALK-related malignancies (ERIA), 1090 Vienna, Austria
- Correspondence: ; Tel.: +49-30-94062863; Fax: +49-30-94063124
| |
Collapse
|
26
|
Abstract
Multiple myeloma is the second most common hematological malignancy in adults, accounting for 2% of all cancer-related deaths in the UK. Current chemotherapy-based regimes are insufficient, as most patients relapse and develop therapy resistance. This review focuses on current novel antibody- and aptamer-based therapies aiming to overcome current therapy limitations, as well as their respective limitations and areas of improvement. The use of computer modeling methods, as a tool to study and improve ligand-receptor alignments for the use of novel therapy development will also be discussed, as it has become a rapid, reliable and comparatively inexpensive method of investigation.
Collapse
|
27
|
Hashmi H, Darwin A, Nishihori T. Therapeutic roles of antibody drug conjugates (ADCs) in relapsed/refractory lymphomas. Hematol Oncol Stem Cell Ther 2021; 16:21-34. [PMID: 36634275 DOI: 10.1016/j.hemonc.2021.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/28/2021] [Accepted: 07/10/2021] [Indexed: 01/18/2023] Open
Abstract
Relapsed or refractory lymphoma is commonly treated with combination chemoimmunotherapy and cellular immunotherapy. Modest response rates and associated toxicities are obstacles to achieving durable remission using traditional cytotoxic chemotherapy, especially in frail patients with advanced disease. Antibody drug conjugates represent a new class of novel targeted agents with significant improvement in therapeutic efficacy in the treatment of lymphomas. Several of these agents, which offer improved targeting, greater potency, and better therapeutic index over traditional chemotherapy, are changing the treatment landscape for lymphomas and other hematological malignancies. Despite the therapeutic potential of these agents, the delivery and release of cytotoxic agents to malignant cells through the combination of a monoclonal antibody, payload, and linker represents a complex design challenge. This article reviews the clinical data on currently available antibody drug conjugates and the ongoing development of novel antibody drug conjugates. Antibody drug conjugates constitute an important armamentarium for treatment of lymphomas and their evolving roles in the treatment spectrum are discussed.
Collapse
Affiliation(s)
- Hamza Hashmi
- Division of Hematology/Oncology, Medical University of South Carolina, United States
| | - Alicia Darwin
- University of South Florida, Morsani College of Medicine, United States
| | - Taiga Nishihori
- Department of Blood & Marrow Transplant and Cellular Immunotherapy (BMT CI), Moffitt Cancer Center, United States
| |
Collapse
|
28
|
Lopes R, Ferreira BV, Caetano J, Barahona F, Carneiro EA, João C. Boosting Immunity against Multiple Myeloma. Cancers (Basel) 2021; 13:1221. [PMID: 33799565 PMCID: PMC8001641 DOI: 10.3390/cancers13061221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the improvement of patient's outcome obtained by the current use of immunomodulatory drugs, proteasome inhibitors or anti-CD38 monoclonal antibodies, multiple myeloma (MM) remains an incurable disease. More recently, the testing in clinical trials of novel drugs such as anti-BCMA CAR-T cells, antibody-drug conjugates or bispecific antibodies broadened the possibility of improving patients' survival. However, thus far, these treatment strategies have not been able to steadily eliminate all malignant cells, and the aim has been to induce a long-term complete response with minimal residual disease (MRD)-negative status. In this sense, approaches that target not only myeloma cells but also the surrounding microenvironment are promising strategies to achieve a sustained MRD negativity with prolonged survival. This review provides an overview of current and future strategies used for immunomodulation of MM focusing on the impact on bone marrow (BM) immunome.
Collapse
Affiliation(s)
- Raquel Lopes
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medicine, University of Lisbon, 1649-028 Lisbon, Portugal
| | - Bruna Velosa Ferreira
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Joana Caetano
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Filipa Barahona
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
| | - Emilie Arnault Carneiro
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
| | - Cristina João
- Lymphoma and Myeloma Research Programme, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal; (R.L.); (B.V.F.); (J.C.); (F.B.); (E.A.C.)
- Faculty of Medical Sciences, NOVA Medical School, 1169-056 Lisbon, Portugal
- Hemato-Oncology Department, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
29
|
Walsh SJ, Bargh JD, Dannheim FM, Hanby AR, Seki H, Counsell AJ, Ou X, Fowler E, Ashman N, Takada Y, Isidro-Llobet A, Parker JS, Carroll JS, Spring DR. Site-selective modification strategies in antibody-drug conjugates. Chem Soc Rev 2021; 50:1305-1353. [PMID: 33290462 DOI: 10.1039/d0cs00310g] [Citation(s) in RCA: 237] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Antibody-drug conjugates (ADCs) harness the highly specific targeting capabilities of an antibody to deliver a cytotoxic payload to specific cell types. They have garnered widespread interest in drug discovery, particularly in oncology, as discrimination between healthy and malignant tissues or cells can be achieved. Nine ADCs have received approval from the US Food and Drug Administration and more than 80 others are currently undergoing clinical investigations for a range of solid tumours and haematological malignancies. Extensive research over the past decade has highlighted the critical nature of the linkage strategy adopted to attach the payload to the antibody. Whilst early generation ADCs were primarily synthesised as heterogeneous mixtures, these were found to have sub-optimal pharmacokinetics, stability, tolerability and/or efficacy. Efforts have now shifted towards generating homogeneous constructs with precise drug loading and predetermined, controlled sites of attachment. Homogeneous ADCs have repeatedly demonstrated superior overall pharmacological profiles compared to their heterogeneous counterparts. A wide range of methods have been developed in the pursuit of homogeneity, comprising chemical or enzymatic methods or a combination thereof to afford precise modification of specific amino acid or sugar residues. In this review, we discuss advances in chemical and enzymatic methods for site-specific antibody modification that result in the generation of homogeneous ADCs.
Collapse
Affiliation(s)
- Stephen J Walsh
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Barilà G, Rizzi R, Zambello R, Musto P. Drug Conjugated and Bispecific Antibodies for Multiple Myeloma: Improving Immunotherapies off the Shelf. Pharmaceuticals (Basel) 2021; 14:40. [PMID: 33430210 PMCID: PMC7825702 DOI: 10.3390/ph14010040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/02/2021] [Accepted: 01/02/2021] [Indexed: 12/13/2022] Open
Abstract
The impressive improvement of overall survival in multiple myeloma (MM) patients in the last years has been mostly related to the availability of new classes of drugs with different mechanisms of action, including proteasome inhibitors (PI), immunomodulating agents (IMiDs), and monoclonal antibodies. However, even with this increased potence of fire, MM still remains an incurable condition, due to clonal selection and evolution of neoplastic clone. This concept underlines the importance of immunotherapy as one of the most relevant tools to try to eradicate the disease. In line with this concept, active and passive immunotherapies represent the most attractive approach to this aim. Antibody-drug conjugate(s) (ADCs) and bispecific antibodies (BsAbs) include two innovative tools in order to limit neoplastic plasma cell growth or even, if used at the time of the best response, to potentially eradicate the tumoral clone. Following their promising results as single agent for advanced disease, at the recent 62nd ASH meeting, encouraging data of several combinations, particularly of ADC(s) with PI or IMiDs, have been reported, suggesting even better results for patients treated earlier. In this paper, we reviewed the characteristics, mechanism of action, and clinical data available for most relevant ADC(s) and BsAbs.
Collapse
Affiliation(s)
- Gregorio Barilà
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padova University School of Medicine, 35121 Padova, Italy;
| | - Rita Rizzi
- Department of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine, 70121 Bari, Italy;
| | - Renato Zambello
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padova University School of Medicine, 35121 Padova, Italy;
| | - Pellegrino Musto
- Department of Emergency and Organ Transplantation, “Aldo Moro” University School of Medicine, 70121 Bari, Italy;
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, 70124 Bari, Italy
| |
Collapse
|
31
|
Dean AQ, Luo S, Twomey JD, Zhang B. Targeting cancer with antibody-drug conjugates: Promises and challenges. MAbs 2021; 13:1951427. [PMID: 34291723 PMCID: PMC8300931 DOI: 10.1080/19420862.2021.1951427] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/03/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are a rapidly expanding class of biotherapeutics that utilize antibodies to selectively deliver cytotoxic drugs to the tumor site. As of May 2021, the U.S. Food and Drug Administration (FDA) has approved ten ADCs, namely Adcetris®, Kadcyla®, Besponsa®, Mylotarg®, Polivy®, Padcev®, Enhertu®, Trodelvy®, Blenrep®, and Zynlonta™ as monotherapy or combinational therapy for breast cancer, urothelial cancer, myeloma, acute leukemia, and lymphoma. In addition, over 80 investigational ADCs are currently being evaluated in approximately 150 active clinical trials. Despite the growing interest in ADCs, challenges remain to expand their therapeutic index (with greater efficacy and less toxicity). Recent advances in the manufacturing technology for the antibody, payload, and linker combined with new bioconjugation platforms and state-of-the-art analytical techniques are helping to shape the future development of ADCs. This review highlights the current status of marketed ADCs and those under clinical investigation with a focus on translational strategies to improve product quality, safety, and efficacy.
Collapse
Affiliation(s)
- Alexis Q. Dean
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Shen Luo
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Julianne D. Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
32
|
Yang Y, Li Y, Gu H, Dong M, Cai Z. Emerging agents and regimens for multiple myeloma. J Hematol Oncol 2020; 13:150. [PMID: 33168044 PMCID: PMC7654052 DOI: 10.1186/s13045-020-00980-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/19/2020] [Indexed: 02/08/2023] Open
Abstract
The outcomes of multiple myeloma (MM) have been improved significantly with the therapies incorporating proteasome inhibitors (PI), immunomodulatory drugs, monoclonal antibodies (MoAb) and stem cell transplantation. However, relapsed and refractory MM (RRMM) remains a major challenge. Novel agents and regimens are under active clinical development. These include new PIs such as ixazomib, marizomib, and oprozomib; new MoAbs such as isatuximab and MOR202; novel epigenetic agent ricolinostat and novel cytokines such as siltuximab. Recently, the first XPO-1 inhibitor, selinexor, was approved for RRMM. BCMA-targeted BiTE, antibody-drug conjugates and CAR-T cells have the potential to revolutionize the therapy for RRMM. In this review, we summarized the latest clinical development of these novel agents and regimens.
Collapse
Affiliation(s)
- Yang Yang
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yi Li
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiyao Gu
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Mengmeng Dong
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
- Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
33
|
Lieser RM, Yur D, Sullivan MO, Chen W. Site-Specific Bioconjugation Approaches for Enhanced Delivery of Protein Therapeutics and Protein Drug Carriers. Bioconjug Chem 2020; 31:2272-2282. [DOI: 10.1021/acs.bioconjchem.0c00456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel M. Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| |
Collapse
|
34
|
Abstract
Introduction: Antibody-drug conjugates (ADC) are a new class of treatment for multiple myeloma (MM) patients, delivering a potent cytotoxic agent directly to the myeloma cell. The target is defined by the specificity of the monoclonal antibody which is linked to the cytotoxic agent. This mechanism of action minimizes bystander cell injury and allows a favorable therapeutic window.Areas covered: This review describes the rationale, pre- and clinical data for ADCs that have been and are currently in development for MM. As the treatment landscape for MM rapidly evolves, the treatment paradigm and a description of novel agents in development including immunotherapies are provided to understand how ADCs may fit in the pathway.Expert opinion: ADCs have a significant potential for the treatment for MM. As they are 'off the shelf' treatments, they can be used across nearly all MM treatment centers and to a wide range of patients. Some ADCs have specific adverse events that may require specialist input to optimally manage. The most clinically advanced ADC is belantamab mafodotin which has demonstrated clinically meaningful responses in patients with heavily pre-treated MM. Additionally, it is being combined with standard of care agents and at earlier lines of treatment.
Collapse
Affiliation(s)
- Annabel McMillan
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| | - Dana Warcel
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| | - Rakesh Popat
- Haematology Department, National Institute for Health Research University College Hospital Clinical Research Facility, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
35
|
Bruins WSC, Zweegman S, Mutis T, van de Donk NWCJ. Targeted Therapy With Immunoconjugates for Multiple Myeloma. Front Immunol 2020; 11:1155. [PMID: 32636838 PMCID: PMC7316960 DOI: 10.3389/fimmu.2020.01155] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
The introduction of proteasome inhibitors (PI) and immunomodulatory drugs (IMiD) has markedly increased the survival of multiple myeloma (MM) patients. Also, the unconjugated monoclonal antibodies (mAb) daratumumab (anti-CD38) and elotuzumab (anti-SLAMF7) have revolutionized MM treatment given their clinical efficacy and safety, illustrating the potential of targeted immunotherapy as a powerful treatment strategy for MM. Nonetheless, most patients eventually develop PI-, IMiD-, and mAb-refractory disease because of the selection of resistant MM clones, which associates with a poor prognosis. Accordingly, these patients remain in urgent need of new therapies with novel mechanisms of action. In this respect, mAbs or mAb fragments can also be utilized as carriers of potent effector moieties to specifically target surface antigens on cells of interest. Such immunoconjugates have the potential to exert anti-MM activity in heavily pretreated patients due to their distinct and pleiotropic mechanisms of action. In addition, the fusion of highly cytotoxic compounds to mAbs decreases the off-target toxicity, thereby improving the therapeutic window. According to the effector moiety, immunoconjugates are classified into antibody-drug conjugates, immunotoxins, immunocytokines, or radioimmunoconjugates. This review will focus on the mechanisms of action, safety and efficacy of several promising immunoconjugates that are under investigation in preclinical and/or clinical MM studies. We will also include a discussion on combination therapy with immunoconjugates, resistance mechanisms, and future developments.
Collapse
Affiliation(s)
- Wassilis S C Bruins
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sonja Zweegman
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tuna Mutis
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Niels W C J van de Donk
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Gauzy-Lazo L, Sassoon I, Brun MP. Advances in Antibody–Drug Conjugate Design: Current Clinical Landscape and Future Innovations. SLAS DISCOVERY 2020; 25:843-868. [DOI: 10.1177/2472555220912955] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The targeted delivery of potent cytotoxic molecules into cancer cells is considered a promising anticancer strategy. The design of clinically effective antibody–drug conjugates (ADCs), in which biologically active drugs are coupled through chemical linkers to monoclonal antibodies, has presented challenges for pharmaceutical researchers. After 30 years of intensive research and development activities, only seven ADCs have been approved for clinical use; two have received fast-track designation and two breakthrough therapy designation from the Food and Drug Administration. There is continued interest in the field, as documented by the growing number of candidates in clinical development. This review aims to summarize the most recent innovations that have been applied to the design of ADCs undergoing early- and late-stage clinical trials. Discovery and rational optimization of new payloads, chemical linkers, and antibody formats have improved the therapeutic index of next-generation ADCs, ultimately resulting in improved clinical benefit for the patients.
Collapse
Affiliation(s)
| | - Ingrid Sassoon
- Immuno-Oncology Therapeutic Area, Sanofi, Vitry-sur-Seine, France
| | | |
Collapse
|
37
|
Cohen AD. Myeloma: next generation immunotherapy. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2019; 2019:266-272. [PMID: 31808859 PMCID: PMC6913481 DOI: 10.1182/hematology.2019000068] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The course of multiple myeloma (MM) from initial diagnosis to a relapsed/refractory state is characterized by acquisition of drug resistance as well as progressive immunologic dysfunction. Despite this, however, a number of novel therapies that work in part or solely via immune stimulation are in development for MM, with promising early clinical results. Several new whole-cell or multiepitope vaccine approaches are demonstrating immunologic efficacy in smoldering MM or as posttherapy consolidation, with trials ongoing to see whether this translates into delayed progression or elimination of minimal residual disease. Programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibition in combination with immunomodulatory drugs demonstrated excessive toxicity in randomized trials; however, antibodies targeting PD-1/PD-L1 and other checkpoint molecules continue to be explored in combination with tumor-targeted antibodies and other T cell-directed therapies. B-cell maturation antigen (BCMA) has emerged as the next big antigen target, with multiple BCMA-specific antibody-drug conjugates (ADCs) and T cell-directed bispecific antibodies/bispecific therapeutic engagers (BiTEs) entering the clinic. In initial trials, the ADC GSK2857916 and the BiTE AMG 420 have demonstrated high response rates in relapsed/refractory patients, with depth and durability of responses that may end up rivaling chimeric antigen receptor T-cell therapies. These agents have unique toxicities that require close monitoring, but they are moving forward in larger registration studies and in combination with standard MM agents. Additional ADCs and bispecific antibodies targeting BCMA and other surface antigens (eg, CD38, CD46, CD48, FcRH5, and G protein-coupled receptor, class C group 5 member D) are moving forward in phase 1 trials and may provide even more options for MM patients.
Collapse
Affiliation(s)
- Adam D Cohen
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
38
|
Song Y, Sun Y, Lei Y, Yang K, Tang R. YAP1 promotes multidrug resistance of small cell lung cancer by CD74-related signaling pathways. Cancer Med 2019; 9:259-268. [PMID: 31692299 PMCID: PMC6943160 DOI: 10.1002/cam4.2668] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/12/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Our previous research found that YAP1 may have a role in multidrug resistance (MDR) in small cell lung cancer (SCLC). However, its underlying mechanism is unknown. METHODS In this study, we investigated the expression of YAP1 using immunohistochemical staining and assessed the relationship between the expression of YAP1 and overall survival in patients with SCLC. We established H69 stable cell lines that overexpressed constitutively active YAP1 and H446 stable cell lines that dominate negative YAP1. We conducted CCK-8, flow cytometric analysis, and in vivo chemosensitivity experiments to evaluate the function of YAP1 in drug sensitivity apoptosis in vitro and in vivo. RESULTS The results indicated that patients with high YAP1 expression have shorter survival rates and more advanced disease stage than those with low YAP1 expression. YAP1 may induce MDR by inhibiting the apoptosis of SCLC. YAP1 induced MDR when YAP1 was hyperactivated, and drug sensitivity increased when YAP1 was inhibited in vitro and in vivo. CD74 was significantly correlated with YAP1 in SCLC samples. Inhibition of CD74 using ISO-1 increased drug sensitivity significantly. CONCLUSIONS The expression of YAP1 is significantly correlated with overall survival and disease stage in patients with SCLC. YAP1 may play an important role in these patients. We were the first to report that YAP1 can induce MDR in SCLC in vitro and in vivo. CD74 may be involved in YAP1-induced MDR.
Collapse
Affiliation(s)
- Yongchun Song
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanqin Sun
- Department of Pathology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yingying Lei
- Department of Oncology, Panyu Maternal and Child Care Service Centre of Guangzhou and Hexian Memorial affiliated hospital of Southern Medical University, Guangzhou, China
| | - Kui Yang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ruixiang Tang
- Department of Oncology Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
39
|
Lim SI. Site-specific bioconjugation and self-assembly technologies for multi-functional biologics: on the road to the clinic. Drug Discov Today 2019; 25:168-176. [PMID: 31610287 DOI: 10.1016/j.drudis.2019.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 01/02/2023]
Abstract
The expanding portfolio of biotherapeutics both in the research and development (R&D) and market sectors is shaping new opportunities towards multifunctional biologics (MFBs). The combination of new or pre-existing therapeutic agents into a single multifunctional format makes it possible to develop new pharmacological actions to significantly improve their efficacy and safety. In this review, I focus on novel platform technologies that are being exploited in the biotech industry to produce MFBs with potential therapeutic benefits that include half-life extension, targeted delivery, T cell engagement, and improved vaccination. In this regard, technologies of key importance are site-specific bioconjugation and self-assembly, which allow homogeneous, defined, and scalable process developments for several MFBs that are advancing towards clinical applications.
Collapse
Affiliation(s)
- Sung In Lim
- Department of Chemical Engineering, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan, Republic of Korea.
| |
Collapse
|