1
|
Ababneh O, Nishizaki D, Kato S, Kurzrock R. Tumor necrosis factor superfamily signaling: life and death in cancer. Cancer Metastasis Rev 2024; 43:1137-1163. [PMID: 39363128 PMCID: PMC11554763 DOI: 10.1007/s10555-024-10206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 10/05/2024]
Abstract
Immune checkpoint inhibitors have shaped the landscape of cancer treatment. However, many patients either do not respond or suffer from later progression. Numerous proteins can control immune system activity, including multiple tumor necrosis factor (TNF) superfamily (TNFSF) and TNF receptor superfamily (TNFRSF) members; these proteins play a complex role in regulating cell survival and death, cellular differentiation, and immune system activity. Notably, TNFSF/TNFRSF molecules may display either pro-tumoral or anti-tumoral activity, or even both, depending on tumor type. Therefore, TNF is a prototype of an enigmatic two-faced mediator in oncogenesis. To date, multiple anti-TNF agents have been approved and/or included in guidelines for treating autoimmune disorders and immune-related toxicities after immune checkpoint blockade for cancer. A confirmed role for the TNFSF/TNFRSF members in treating cancer has proven more elusive. In this review, we highlight the cancer-relevant TNFSF/TNFRSF family members, focusing on the death domain-containing and co-stimulation members and their signaling pathways, as well as their complicated role in the life and death of cancer cells.
Collapse
Affiliation(s)
- Obada Ababneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium, Paris, France.
- Department of Medicine, MCW Cancer Center, Milwaukee, WI, USA.
- Department of Oncology, University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
2
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2024:10.1038/s41577-024-01091-9. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Tran B, Voskoboynik M, Bendell J, Gutierrez M, Lemech C, Day D, Frentzas S, Garrido-Laguna I, Standifer N, Wang F, Ferte C, Wang Y, Das M, Carneiro BA. A phase 1 study of the CD40 agonist MEDI5083 in combination with durvalumab in patients with advanced solid tumors. Immunotherapy 2024; 16:759-774. [PMID: 39264730 PMCID: PMC11421296 DOI: 10.1080/1750743x.2024.2359359] [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: 12/12/2023] [Accepted: 05/21/2024] [Indexed: 09/14/2024] Open
Abstract
Aim: This first-in-human study evaluated safety and efficacy of CD40 agonist MEDI5083 with durvalumab in patients with advanced solid tumors.Methods: Patients received MEDI5083 (3-7.5 mg subcutaneously every 2 weeks × 4 doses) and durvalumab (1500 mg every 4 weeks) either sequentially (N = 29) or concurrently (N = 9). Primary end point was safety; secondary end points included efficacy.Results: Thirty-eight patients received treatment. Most common adverse events (AEs) were injection-site reaction (ISR; sequential: 86%; concurrent: 100%), fatigue (41%; 33%), nausea (20.7%; 55.6%) and decreased appetite (24.1%; 33.3%). Nine patients had MEDI5083-related grade ≥3 AEs with ISR being the most common. Two patients experienced dose limiting toxicities (ISR). One death occurred due to a MEDI5083-related AE. MEDI5083 maximum tolerated dose was 5 mg. Objective response rate was 2.8% (1 partial response and 11 stable disease).Conclusion: MEDI5083 toxicity profile limits its further development.
Collapse
Affiliation(s)
- Ben Tran
- Peter MacCallum Cancer Centre, Melbourne, 8006, Australia
| | - Mark Voskoboynik
- Nucleus Network, Melbourne, 3004, Australia
- Monash University, Melbourne, 3004, Australia
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN 37203, USA
| | | | - Charlotte Lemech
- Scientia Clinical Research, Randwick, 2031, Australia
- The University of New South Wales, Sydney, 2052, Australia
| | - Daphne Day
- Monash University, Melbourne, 3004, Australia
- Monash Medical Centre, Clayton, 3800, Australia
| | - Sophia Frentzas
- Monash University, Melbourne, 3004, Australia
- Monash Medical Centre, Clayton, 3800, Australia
| | | | - Nathan Standifer
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Fujun Wang
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD 20878,USA
| | | | - Yue Wang
- AstraZeneca, Gaithersburg, MD 20878,USA
| | | | - Benedito A Carneiro
- Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, RI 02903,USA
| |
Collapse
|
4
|
Zhao Y, Hou J, Liu Y, Xu J, Guo Y. An arabinose-rich heteropolysaccharide isolated from Belamcanda chinensis (L.) DC treats liver cancer by targeting FAK and activating CD40. Carbohydr Polym 2024; 331:121831. [PMID: 38388048 DOI: 10.1016/j.carbpol.2024.121831] [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: 08/12/2023] [Revised: 01/12/2024] [Accepted: 01/13/2024] [Indexed: 02/24/2024]
Abstract
An undisclosed polysaccharide, BCP80-2, was isolated from Belamcanda chinensis (L.) DC. Structural investigation revealed that BCP80-2 consists of ten monosaccharide residues including t-α-Araf-(1→, →3,5)-α-Araf-(1→, →5)-α-Araf-(1→, →4)-β-Xylp-(1→, →3)-α-Rhap-(1→, →4)-β-Manp-(1→, t-β-Glcp-(1→, →6)-α-Glcp-(1→, t-β-Galp-(1→, and→3)-α-Galp-(1→. In vivo activity assays showed that BCP80-2 significantly suppressed neoplasmic growth, metastasis, and angiogenesis in zebrafish. Mechanistic studies have shown that BCP80-2 inhibited cell migration of HepG2 cells by suppressing the FAK signaling pathway. Moreover, BCP80-2 also activated immunomodulation and upregulated the secretion of co-stimulatory molecules CD40, CD86, CD80, and MHC-II. In conclusion, BCP80-2 inhibited tumor progression by targeting the FAK signaling pathway and activating CD40-induced adaptive immunity.
Collapse
Affiliation(s)
- Yinan Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Jiantong Hou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yuhui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China.
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China.
| |
Collapse
|
5
|
Andersson H, Nyesiga B, Hermodsson T, Enell Smith K, Hägerbrand K, Lindstedt M, Ellmark P. Next-generation CD40 agonists for cancer immunotherapy. Expert Opin Biol Ther 2024; 24:351-363. [PMID: 38764393 DOI: 10.1080/14712598.2024.2357714] [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: 02/16/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION There is a need for new therapies that can enhance response rates and broaden the number of cancer indications where immunotherapies provide clinical benefit. CD40 targeting therapies provide an opportunity to meet this need by promoting priming of tumor-specific T cells and reverting the suppressive tumor microenvironment. This is supported by emerging clinical evidence demonstrating the benefits of immunotherapy with CD40 antibodies in combination with standard of care chemotherapy. AREAS COVERED This review is focused on the coming wave of next-generation CD40 agonists aiming to improve efficacy and safety, using new approaches and formats beyond monospecific antibodies. Further, the current understanding of the role of different CD40 expressing immune cell populations in the tumor microenvironment is reviewed. EXPERT OPINION There are multiple promising next-generation approaches beyond monospecific antibodies targeting CD40 in immuno-oncology. Enhancing efficacy is the most important driver for this development, and approaches that maximize the ability of CD40 to both remodel the tumor microenvironment and boost the anti-tumor T cell response provide great opportunities to benefit cancer patients. Enhanced understanding of the role of different CD40 expressing immune cells in the tumor microenvironment may facilitate more efficient clinical development of these compounds.
Collapse
Affiliation(s)
- Hampus Andersson
- Alligator Bioscience, Alligator Bioscience AB, Lund, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Barnabas Nyesiga
- Alligator Bioscience, Alligator Bioscience AB, Lund, Sweden
- Department of Biomedical Science, Malmö University, Malmö, Sweden
| | - Tova Hermodsson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | | | - Malin Lindstedt
- Alligator Bioscience, Alligator Bioscience AB, Lund, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Peter Ellmark
- Alligator Bioscience, Alligator Bioscience AB, Lund, Sweden
- Department of Immunotechnology, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Beckmann K, Reitinger C, Yan X, Carle A, Blümle E, Jurkschat N, Paulmann C, Prassl S, Kazandjian LV, Loré K, Nimmerjahn F, Fischer S. Fcγ-Receptor-Independent Controlled Activation of CD40 Canonical Signaling by Novel Therapeutic Antibodies for Cancer Therapy. Antibodies (Basel) 2024; 13:31. [PMID: 38651411 PMCID: PMC11036229 DOI: 10.3390/antib13020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/04/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
The activation of CD40-mediated signaling in antigen-presenting cells is a promising therapeutic strategy to promote immune responses against tumors. Most agonistic anti-CD40 antibodies currently in development require the Fcγ-receptor (FcγR)-mediated crosslinking of CD40 molecules for a meaningful activation of CD40 signaling but have limitations due to dose-limiting toxicities. Here we describe the identification of CD40 antibodies which strongly stimulate antigen-presenting cells in an entirely FcγR-independent manner. These Fc-silenced anti-CD40 antibodies induce an efficient upregulation of costimulatory receptors and cytokine release by dendritic cells. Finally, the most active identified anti-CD40 antibody shows activity in humanized mice. More importantly, there are no signs of obvious toxicities. These studies thus demonstrate the potent activation of antigen-presenting cells with anti-CD40 antibodies lacking FcγR-binding activity and open the possibility for an efficacious and safe combination therapy for cancer patients.
Collapse
Affiliation(s)
| | - Carmen Reitinger
- Division of Genetics, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Xianglei Yan
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Visionsgatan 4, BioClinicum J7:30, 171 64, Stockholm, Sweden
- Center of Molecular Medicine, 171 76, Stockholm, Sweden
| | - Anna Carle
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | - Eva Blümle
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | | | | | - Sandra Prassl
- Biontech SE, Forstenrieder Str. 8-14, 82061 Neuried, Germany
| | | | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Visionsgatan 4, BioClinicum J7:30, 171 64, Stockholm, Sweden
- Center of Molecular Medicine, 171 76, Stockholm, Sweden
| | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, 91058 Erlangen, Germany
- FAU Profile Centre Immunomedicine, 91054 Erlangen, Germany
| | | |
Collapse
|
7
|
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024; 23:301-319. [PMID: 38448606 DOI: 10.1038/s41573-024-00896-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
8
|
Zhou Y, Richmond A, Yan C. Harnessing the potential of CD40 agonism in cancer therapy. Cytokine Growth Factor Rev 2024; 75:40-56. [PMID: 38102001 PMCID: PMC10922420 DOI: 10.1016/j.cytogfr.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily of receptors expressed on a variety of cell types. The CD40-CD40L interaction gives rise to many immune events, including the licensing of dendritic cells to activate CD8+ effector T cells, as well as the facilitation of B cell activation, proliferation, and differentiation. In malignant cells, the expression of CD40 varies among cancer types, mediating cellular proliferation, apoptosis, survival and the secretion of cytokines and chemokines. Agonistic human anti-CD40 antibodies are emerging as an option for cancer treatment, and early-phase clinical trials explored its monotherapy or combination with radiotherapy, chemotherapy, immune checkpoint blockade, and other immunomodulatory approaches. In this review, we present the current understanding of the mechanism of action for CD40, along with results from the clinical development of agonistic human CD40 antibodies in cancer treatment (selicrelumab, CDX-1140, APX005M, mitazalimab, 2141-V11, SEA-CD40, LVGN7409, and bispecific antibodies). This review also examines the safety profile of CD40 agonists in both preclinical and clinical settings, highlighting optimized dosage levels, potential adverse effects, and strategies to mitigate them.
Collapse
Affiliation(s)
- Yang Zhou
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Chi Yan
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA.
| |
Collapse
|
9
|
Romei MG, Leonard B, Katz ZB, Le D, Yang Y, Day ES, Koo CW, Sharma P, Bevers Iii J, Kim I, Dai H, Farahi F, Lin M, Shaw AS, Nakamura G, Sockolosky JT, Lazar GA. i-shaped antibody engineering enables conformational tuning of biotherapeutic receptor agonists. Nat Commun 2024; 15:642. [PMID: 38245524 PMCID: PMC10799922 DOI: 10.1038/s41467-024-44985-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 01/11/2024] [Indexed: 01/22/2024] Open
Abstract
The ability to leverage antibodies to agonize disease relevant biological pathways has tremendous potential for clinical investigation. Yet while antibodies have been successful as antagonists, immune mediators, and targeting agents, they are not readily effective at recapitulating the biology of natural ligands. Among the important determinants of antibody agonist activity is the geometry of target receptor engagement. Here, we describe an engineering approach inspired by a naturally occurring Fab-Fab homotypic interaction that constrains IgG in a unique i-shaped conformation. i-shaped antibody (iAb) engineering enables potent intrinsic agonism of five tumor necrosis factor receptor superfamily (TNFRSF) targets. When applied to bispecific antibodies against the heterodimeric IL-2 receptor pair, constrained bispecific IgG formats recapitulate IL-2 agonist activity. iAb engineering provides a tool to tune agonist antibody function and this work provides a framework for the development of intrinsic antibody agonists with the potential for generalization across broad receptor classes.
Collapse
Affiliation(s)
- Matthew G Romei
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Brandon Leonard
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Zachary B Katz
- Department of Research Biology, Genentech Inc., South San Francisco, CA, USA
| | - Daniel Le
- Department of Microchemistry, Proteomic, Lipidomics, and Next Generation Sequencing, Genentech Inc., South San Francisco, CA, USA
| | - Yanli Yang
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Eric S Day
- Department of Pharma Technical Development, Genentech Inc., South San Francisco, CA, USA
| | - Christopher W Koo
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Preeti Sharma
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Jack Bevers Iii
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Ingrid Kim
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Huiguang Dai
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - Farzam Farahi
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | - May Lin
- Department of Protein Chemistry, Genentech Inc., South San Francisco, CA, USA
| | - Andrey S Shaw
- Department of Research Biology, Genentech Inc., South San Francisco, CA, USA
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA
| | | | - Greg A Lazar
- Department of Antibody Engineering, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
10
|
Andersson H, Sobti A, Jimenez DG, de Coaña YP, Ambarkhane SV, Hägerbrand K, Smith KE, Lindstedt M, Ellmark P. Early Pharmacodynamic Changes Measured Using RNA Sequencing of Peripheral Blood from Patients in a Phase I Study with Mitazalimab, a Potent CD40 Agonistic Monoclonal Antibody. Cells 2023; 12:2365. [PMID: 37830579 PMCID: PMC10572020 DOI: 10.3390/cells12192365] [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: 08/23/2023] [Revised: 09/18/2023] [Accepted: 09/22/2023] [Indexed: 10/14/2023] Open
Abstract
CD40-targeting therapies can enhance the dendritic cell priming of tumor-specific T cells and repolarize intratumoral macrophages to alleviate the tumoral immunosuppressive environment and remodel the extracellular matrix. Mitazalimab is a potent agonistic CD40 monoclonal IgG1 antibody currently under clinical development. This study used RNA sequencing of blood samples from a subset of patients from a Phase I trial with mitazalimab (NCT02829099) to assess peripheral pharmacodynamic activity. We found that mitazalimab induced transient peripheral transcriptomic alterations (at 600 µg/kg and 900 µg/kg dose administered intravenously), which mainly were attributed to immune activation. In particular, the transcriptomic alterations showed a reduction in effector cells (e.g., CD8+ T cells and natural killer cells) and B cells peripherally with the remaining cells (e.g., dendritic cells, monocytes, B cells, and natural killer cells) showing transcription profiles consistent with activation. Lastly, distinct patient subgroups based on the pattern of transcriptomic alterations could be identified. In summary, the data presented herein reinforce the anticipated mode of action of mitazalimab and support its ongoing clinical development.
Collapse
Affiliation(s)
- Hampus Andersson
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
- Department of Immunotechnology, Lund University, 223 81 Lund, Sweden
| | - Aastha Sobti
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
| | - David Gomez Jimenez
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
| | - Yago Pico de Coaña
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
| | | | - Karin Hägerbrand
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
| | - Karin Enell Smith
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
| | - Malin Lindstedt
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
- Department of Immunotechnology, Lund University, 223 81 Lund, Sweden
| | - Peter Ellmark
- Alligator Bioscience AB, 223 81 Lund, Sweden; (H.A.); (A.S.); (D.G.J.); (Y.P.d.C.); (M.L.)
- Department of Immunotechnology, Lund University, 223 81 Lund, Sweden
| |
Collapse
|
11
|
Babar Q, Saeed A, Tabish TA, Sarwar M, Thorat ND. Targeting the tumor microenvironment: Potential strategy for cancer therapeutics. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166746. [PMID: 37160171 DOI: 10.1016/j.bbadis.2023.166746] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Cellular and stromal components including tumor cells, immune cells, mesenchymal cells, cancer-linked fibroblasts, and extracellular matrix, constituent tumor microenvironment (TME). TME plays a crucial role in reprogramming tumor initiation, uncontrolled proliferation, invasion and metastasis as well as response to therapeutic modalities. In recent years targeting the TME has developed as a potential strategy for treatment of cancer because of its life-threatening functions in restricting tumor development and modulating responses to standard-of-care medicines. Cold atmospheric plasma, oncolytic viral therapy, bacterial therapy, nano-vaccine, and repurposed pharmaceuticals with combination therapy, antiangiogenic drugs, and immunotherapies are among the most effective therapies directed by TME that have either been clinically authorized or are currently being studied. This article discusses above-mentioned therapies in light of targeting TME. We also cover problems related to the TME-targeted therapies, as well as future insights and practical uses in this rapidly growing field.
Collapse
Affiliation(s)
- Quratulain Babar
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Ayesha Saeed
- Department of Biochemistry Government College University, Faisalabad, Pakistan
| | - Tanveer A Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Mohsin Sarwar
- Department of Biochemistry University of Management and Technology, Lahore, Pakistan
| | - Nanasaheb D Thorat
- Department of Physics, Bernal Institute, Castletroy, Limerick V94T9PX, Ireland; Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, Medical Sciences Division, University of Oxford, Oxford OX3 9DU, United Kingdom; Limerick Digital Cancer Research Centre (LDCRC) University of Limerick, Castletroy, Limerick V94T9PX, Ireland.
| |
Collapse
|
12
|
Manso T, Kushwaha A, Abdollahi N, Duroux P, Giudicelli V, Kossida S. Mechanisms of action of monoclonal antibodies in oncology integrated in IMGT/mAb-DB. Front Immunol 2023; 14:1129323. [PMID: 37215135 PMCID: PMC10196129 DOI: 10.3389/fimmu.2023.1129323] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/07/2023] [Indexed: 05/24/2023] Open
Abstract
Background Cancer cells activate different immune checkpoint (IC) pathways in order to evade immunosurveillance. Immunotherapies involving ICs either block or stimulate these pathways and enhance the efficiency of the immune system to recognize and attack cancer cells. In this way, the development of monoclonal antibodies (mAbs) targeting ICs has significant success in cancer treatment. Recently, a systematic description of the mechanisms of action (MOA) of the mAbs has been introduced in IMGT/mAb-DB, the IMGT® database dedicated to mAbs for therapeutic applications. The characterization of these antibodies provides a comprehensive understanding of how mAbs work in cancer. Methods In depth biocuration taking advantage of the abundant literature data as well as amino acid sequence analyses from mAbs managed in IMGT/2Dstructure-DB, the IMGT® protein database, allowed to define a standardized and consistent description of the MOA of mAbs targeting immune checkpoints in cancer therapy. Results A fine description and a standardized graphical representation of the MOA of selected mAbs are integrated within IMGT/mAb-DB highlighting two main mechanisms in cancer immunotherapy, either Blocking or Agonist. In both cases, the mAbs enhance cytotoxic T lymphocyte (CTL)-mediated anti-tumor immune response (Immunostimulant effect) against tumor cells. On the one hand, mAbs targeting co-inhibitory receptors may have a functional Fc region to increase anti-tumor activity by effector properties that deplete Treg cells (Fc-effector function effect) or may have limited FcγR binding to prevent Teff cells depletion and reduce adverse events. On the other hand, agonist mAbs targeting co-stimulatory receptors may bind to FcγRs, resulting in antibody crosslinking (FcγR crosslinking effect) and substantial agonism. Conclusion In IMGT/mAb-DB, mAbs for cancer therapy are characterized by their chains, domains and sequence and by several therapeutic metadata, including their MOA. MOAs were recently included as a search criterion to query the database. IMGT® is continuing standardized work to describe the MOA of mAbs targeting additional immune checkpoints and novel molecules in cancer therapy, as well as expanding this study to other clinical domains.
Collapse
|
13
|
Zhang E, Ding C, Li S, Zhou X, Aikemu B, Fan X, Sun J, Zheng M, Yang X. Roles and mechanisms of tumour-infiltrating B cells in human cancer: a new force in immunotherapy. Biomark Res 2023; 11:28. [PMID: 36890557 PMCID: PMC9997025 DOI: 10.1186/s40364-023-00460-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/28/2023] [Indexed: 03/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting PD-1 or PD-L1 have emerged as a revolutionary treatment strategy for human cancer patients. However, as the response rate to ICI therapy varies widely among different types of tumours, we are beginning to gain insight into the mechanisms as well as biomarkers of therapeutic response and resistance. Numerous studies have highlighted the dominant role of cytotoxic T cells in determining the treatment response to ICIs. Empowered by recent technical advances, such as single-cell sequencing, tumour-infiltrating B cells have been identified as a key regulator in several solid tumours by affecting tumour progression and the response to ICIs. In the current review, we summarized recent advances regarding the role and underlying mechanisms of B cells in human cancer and therapy. Some studies have shown that B-cell abundance in cancer is positively associated with favourable clinical outcomes, while others have indicated that they are tumour-promoting, implying that the biological function of B cells is a complex landscape. The molecular mechanisms involved multiple aspects of the functions of B cells, including the activation of CD8+ T cells, the secretion of antibodies and cytokines, and the facilitation of the antigen presentation process. In addition, other crucial mechanisms, such as the functions of regulatory B cells (Bregs) and plasma cells, are discussed. Here, by summarizing the advances and dilemmas of recent studies, we depicted the current landscape of B cells in cancers and paved the way for future research in this field.
Collapse
Affiliation(s)
- Enkui Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengsheng Ding
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xueliang Zhou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Batuer Aikemu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaodong Fan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Department of General Surgery & Carson International Cancer Research Center, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China.
| | - Xiao Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China. .,Department of General Surgery & Carson International Cancer Research Center, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy, Shenzhen, 518055, China.
| |
Collapse
|
14
|
Hägerbrand K, Varas L, Deronic A, Nyesiga B, Sundstedt A, Ljung L, Sakellariou C, Werchau D, Thagesson M, Gomez Jimenez D, Greiff L, Celander M, Smedenfors K, Rosén A, Bölükbas D, Carlsson F, Levin M, Säll A, von Schantz L, Lindstedt M, Ellmark P. Bispecific antibodies targeting CD40 and tumor-associated antigens promote cross-priming of T cells resulting in an antitumor response superior to monospecific antibodies. J Immunother Cancer 2022; 10:jitc-2022-005018. [PMID: 36323431 PMCID: PMC9660648 DOI: 10.1136/jitc-2022-005018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Indications with poor T-cell infiltration or deficiencies in T-cell priming and associated unresponsiveness to established immunotherapies represent an unmet medical need in oncology. CD40-targeting therapies designed to enhance antigen presentation, generate new tumor-specific T cells, and activate tumor-infiltrating myeloid cells to remodel the tumor microenvironment, represent a promising opportunity to meet this need. In this study, we present the first in vivo data supporting a role for tumor-associated antigen (TAA)-mediated uptake and cross-presentation of tumor antigens to enhance tumor-specific T-cell priming using CD40×TAA bispecific antibodies, a concept we named Neo-X-Prime. METHODS Bispecific antibodies targeting CD40 and either of two cell-surface expressed TAA, carcinoembryonic antigen-related cell adhesion molecule 5 (CEA) or epithelial cell adhesion molecule (EpCAM), were developed in a tetravalent format. TAA-conditional CD40 agonism, activation of tumor-infiltrating immune cells, antitumor efficacy and the role of delivery of tumor-derived material such as extracellular vesicles, tumor debris and exosomes by the CD40×TAA bispecific antibodies were demonstrated in vitro using primary human and murine cells and in vivo using human CD40 transgenic mice with different tumor models. RESULTS The results showed that the CD40×TAA bispecific antibodies induced TAA-conditional CD40 activation both in vitro and in vivo. Further, it was demonstrated in vitro that they induced clustering of tumor debris and CD40-expressing cells in a dose-dependent manner and superior T-cell priming when added to dendritic cells (DC), ovalbumin (OVA)-specific T cells and OVA-containing tumor debris or exosomes. The antitumor activity of the Neo-X-Prime bispecific antibodies was demonstrated to be significantly superior to the monospecific CD40 antibody, and the resulting T-cell dependent antitumor immunity was directed to tumor antigens other than the TAA used for targeting (EpCAM). CONCLUSIONS The data presented herein support the hypothesis that CD40×TAA bispecific antibodies can engage tumor-derived vesicles containing tumor neoantigens to myeloid cells such as DCs resulting in an improved DC-mediated cross-priming of tumor-specific CD8+ T cells. Thus, this principle may offer therapeutics strategies to enhance tumor-specific T-cell immunity and associated clinical benefit in indications characterized by poor T-cell infiltration or deficiencies in T-cell priming.
Collapse
Affiliation(s)
| | - Laura Varas
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Adnan Deronic
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | - Lill Ljung
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | - Mia Thagesson
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | - Lennart Greiff
- Department of ORL, Head & Neck Surgery, Skåne University Hospital, Lund, Sweden
| | - Mona Celander
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | - Anna Rosén
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | | | - Mattias Levin
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | - Anna Säll
- Alligator Bioscience AB, Medicon Village, Lund, Sweden
| | | | - Malin Lindstedt
- Alligator Bioscience AB, Medicon Village, Lund, Sweden,Department of Immunotechnology, Lund University, Lund, Sweden
| | - Peter Ellmark
- Alligator Bioscience AB, Medicon Village, Lund, Sweden,Department of Immunotechnology, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Liu L, Wu Y, Ye K, Cai M, Zhuang G, Wang J. Antibody-Targeted TNFRSF Activation for Cancer Immunotherapy: The Role of FcγRIIB Cross-Linking. Front Pharmacol 2022; 13:924197. [PMID: 35865955 PMCID: PMC9295861 DOI: 10.3389/fphar.2022.924197] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/30/2022] [Indexed: 12/19/2022] Open
Abstract
Co-stimulation signaling in various types of immune cells modulates immune responses in physiology and disease. Tumor necrosis factor receptor superfamily (TNFRSF) members such as CD40, OX40 and CD137/4-1BB are expressed on myeloid cells and/or lymphocytes, and they regulate antigen presentation and adaptive immune activities. TNFRSF agonistic antibodies have been evaluated extensively in preclinical models, and the robust antitumor immune responses and efficacy have encouraged continued clinical investigations for the last two decades. However, balancing the toxicities and efficacy of TNFRSF agonistic antibodies remains a major challenge in the clinical development. Insights into the co-stimulation signaling biology, antibody structural roles and their functionality in immuno-oncology are guiding new advancement of this field. Leveraging the interactions between antibodies and the inhibitory Fc receptor FcγRIIB to optimize co-stimulation agonistic activities dependent on FcγRIIB cross-linking selectively in tumor microenvironment represents the current frontier, which also includes cross-linking through tumor antigen binding with bispecific antibodies. In this review, we will summarize the immunological roles of TNFRSF members and current clinical studies of TNFRSF agonistic antibodies. We will also cover the contribution of different IgG structure domains to these agonistic activities, with a focus on the role of FcγRIIB in TNFRSF cross-linking and clustering bridged by agonistic antibodies. We will review and discuss several Fc-engineering approaches to optimize Fc binding ability to FcγRIIB in the context of proper Fab and the epitope, including a cross-linking antibody (xLinkAb) model and its application in developing TNFRSF agonistic antibodies with improved efficacy and safety for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Yi Wu
- Lyvgen Biopharma, Shanghai, China
| | - Kaiyan Ye
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meichun Cai
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglei Zhuang
- State Key Laboratory of Oncogenes and Related Genes, Department of Obstetrics and Gynecology, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Gynecologic Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | |
Collapse
|
16
|
To N, Evans RPT, Pearce H, Kamarajah SK, Moss P, Griffiths EA. Current and Future Immunotherapy-Based Treatments for Oesophageal Cancers. Cancers (Basel) 2022; 14:3104. [PMID: 35804876 PMCID: PMC9265112 DOI: 10.3390/cancers14133104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Oesophageal cancer is a disease that causes significant morbidity and mortality worldwide, and the prognosis of this condition has hardly improved in the past few years. Standard treatment includes a combination of chemotherapy, radiotherapy and surgery; however, only a proportion of patients go on to treatment intended to cure the disease due to the late presentation of this disease. New treatment options are of utmost importance, and immunotherapy is a new option that has the potential to transform the landscape of this disease. This treatment is developed to act on the changes within the immune system caused by cancer, including checkpoint inhibitors, which have recently shown great promise in the treatment of this disease and have recently been included in the adjuvant treatment of oesophageal cancer in many countries worldwide. This review will outline the mechanisms by which cancer evades the immune system in those diagnosed with oesophageal cancer and will summarize current and ongoing trials that focus on the use of our own immune system to combat disease.
Collapse
Affiliation(s)
- Natalie To
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Richard P. T. Evans
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Sivesh K. Kamarajah
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Ewen A. Griffiths
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2SY, UK
| |
Collapse
|
17
|
Rigamonti N, Veitonmäki N, Domke C, Barsin S, Jetzer S, Abdelmotaleb O, Bessey R, Lekishvili T, Malvezzi F, Gachechiladze M, Behe M, Levitsky V, Trail PA. A multispecific anti-CD40 DARPin® construct induces tumor-selective CD40 activation and tumor regression. Cancer Immunol Res 2022; 10:626-640. [PMID: 35319751 DOI: 10.1158/2326-6066.cir-21-0553] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/20/2021] [Accepted: 03/01/2022] [Indexed: 11/16/2022]
Abstract
The CD40 receptor is an attractive target for cancer immunotherapy. Although a modest pharmacodynamic effect is seen in patients following administration of CD40-targeting monoclonal antibodies (mAb), the doses that could be safely administered do not result in a meaningful clinical response, most likely due to the limited therapeutic window associated with systemic CD40 activation. To overcome this issue, we developed a multispecific DARPin® construct, α-FAPxCD40, which has conditional activity at the site of disease. α-FAPxCD40 activation of CD40 depends on binding to fibroblast activation protein (FAP), a cell surface protease overexpressed in the stroma of solid tumors. In vitro studies demonstrated that α-FAPxCD40 potently activates human antigen-presenting cells in the presence, but not in the absence, of FAP-positive cells. After intravenous injection, a murine surrogate construct (α-mFAPxCD40) accumulated in FAP-positive tumors, elicited rejection of 88% of these tumors and induced memory anti-tumor immunity. Importantly, in contrast to the mouse anti-CD40 tested in parallel, the in vivo anti-tumor activity of α-mFAPxCD40 was neither associated with elevated blood cytokines nor with hepatotoxicity, both of which contribute to the clinical dose-limiting toxicities of several CD40 mAb. This study demonstrates that α-(m)FAPxCD40 engages CD40 in an FAP-restricted manner leading to tumor eradication without signs of peripheral toxicity. This distinct preclinical profile indicates that a favorable therapeutic index may be achieved in humans. It further supports the development of α-FAPxCD40, currently tested in a first-in-human clinical study in patients with solid tumors (NCT05098405).
Collapse
Affiliation(s)
| | | | - Clara Domke
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | - Sophie Barsin
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | - Sarah Jetzer
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | - Ralph Bessey
- Molecular Partners AG, Zurich-Schlieren, Switzerland
| | | | | | | | - Martin Behe
- Paul Scherrer Institute, Villigen, Switzerland
| | | | | |
Collapse
|
18
|
Aschmoneit N, Kocher K, Siegemund M, Lutz MS, Kühl L, Seifert O, Kontermann RE. Fc-based Duokines: dual-acting costimulatory molecules comprising TNFSF ligands in the single-chain format fused to a heterodimerizing Fc (scDk-Fc). Oncoimmunology 2022; 11:2028961. [PMID: 35083097 PMCID: PMC8786347 DOI: 10.1080/2162402x.2022.2028961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Targeting costimulatory receptors of the tumor necrosis factor superfamily (TNFSF) to activate T-cells and promote anti-tumor T-cell function have emerged as a promising strategy in cancer immunotherapy. Previous studies have shown that combining two different members of the TNFSF resulted in dual-acting costimulatory molecules with the ability to activate two different receptors either on the same cell or on different cell types. To achieve prolonged plasma half-life and extended drug disposition, we have developed novel dual-acting molecules by fusing single-chain ligands of the TNFSF to heterodimerizing Fc chains (scDuokine-Fc, scDk-Fc). Incorporating costimulatory ligands of the TNF superfamily into a scDk-Fc molecule resulted in enhanced T-cell proliferation translating in an increased anti-tumor activity in combination with a primary T-cell-activating bispecific antibody. Our data show that the scDk-Fc molecules are potent immune-stimulatory molecules that are able to enhance T-cell mediated anti-tumor responses.
Collapse
Affiliation(s)
- Nadine Aschmoneit
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Katharina Kocher
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Martin Siegemund
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Martina S. Lutz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Lennart Kühl
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Oliver Seifert
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Roland E. Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| |
Collapse
|