1
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
2
|
Mortaheb S, Pezeshki PS, Rezaei N. Bispecific therapeutics: a state-of-the-art review on the combination of immune checkpoint inhibition with costimulatory and non-checkpoint targeted therapy. Expert Opin Biol Ther 2024; 24:1335-1351. [PMID: 39503381 DOI: 10.1080/14712598.2024.2426636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have revolutionized the field of cancer immunotherapy and have enhanced the survival of patients with malignant tumors. However, the overall efficacy of ICIs remains unsatisfactory and is faced with two major concerns of resistance development and occurrence of immune-related adverse events (irAEs). Bispecific antibodies (bsAbs) have emerged as promising strategies with unique mechanisms of action to achieve a better efficacy and safety than monoclonal antibodies (mAbs) or even their combination. BsAbs along with other bispecific platforms such as bispecific fusion proteins, nanobodies, and CAR-T cells may help to avoid development of resistance and reduce irAEs caused by on-target/off-tumor binding effects of mAbs. AREAS COVERED A literature search was performed using PubMed for English-language articles to provide a comprehensive overview of preclinical and clinical studies on bsAbs specified for both immune checkpoints and non-checkpoint molecules as a well-enhanced class of therapeutics. EXPERT OPINION Identifying suitable targets and selecting effective engineering platforms enhance the potential of bsAbs to address the challenges associated with conventional therapies such as ICIs, positioning them as a promising class of therapeutics in the landscape of cancer immunotherapy.
Collapse
Affiliation(s)
- Samin Mortaheb
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parmida Sadat Pezeshki
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Rojas-Salazar Y, Gómez-Montañez E, Rojas-Salazar J, de Anda-Jáuregui G, Hernández-Lemus E. Potential Drug Synergy Through the ERBB2 Pathway in HER2+ Breast Tumors. Int J Mol Sci 2024; 25:12840. [PMID: 39684551 DOI: 10.3390/ijms252312840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
HER2-positive (HER2+) breast cancer is characterized by the overexpression of the ERBB2 (HER2) gene, which promotes aggressive tumor growth and poor prognosis. Targeting the ERBB2 pathway with single-agent therapies has shown limited efficacy due to resistance mechanisms and the complexity of gene interactions within the tumor microenvironment. This study aims to explore potential drug synergies by analyzing gene-drug interactions and combination therapies that target the ERBB2 pathway in HER2+ breast tumors. Using gene co-expression network analysis, we identified 23 metabolic pathways with significant cross-linking of gene interactions, including those involving EGFR tyrosine kinase inhibitors, PI3K, mTOR, and others. We visualized these interactions using Cytoscape to generate individual and combined drug-gene networks, focusing on frequently used drugs such as Erlotinib, Gefitinib, Lapatinib, and Cetuximab. Individual networks highlighted the direct effects of these drugs on their target genes and neighboring genes within the ERBB2 pathway. Combined drug networks, such as those for Cetuximab with Lapatinib, Cetuximab with Erlotinib, and Erlotinib with Lapatinib, revealed potential synergies that could enhance therapeutic efficacy by simultaneously influencing multiple genes and pathways. Our findings suggest that a network-based approach to analyzing drug combinations provides valuable insights into the molecular mechanisms of HER2+ breast cancer and offers promising strategies for overcoming drug resistance and improving treatment outcomes.
Collapse
Affiliation(s)
- Yareli Rojas-Salazar
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Emiliano Gómez-Montañez
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Jorge Rojas-Salazar
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Guillermo de Anda-Jáuregui
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Investigadores e Investigadoras por Mexico Program, Conahcyt, Mexico City 03940, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City 14610, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
4
|
Zhang L, Yan Y, Gao Y, Chen Y, Yu J, Ren N, Sun L. Antibody-drug conjugates and immune checkpoint inhibitors in cancer treatment: a systematic review and meta-analysis. Sci Rep 2024; 14:22357. [PMID: 39333227 PMCID: PMC11436769 DOI: 10.1038/s41598-024-68311-z] [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: 01/28/2024] [Accepted: 07/22/2024] [Indexed: 09/29/2024] Open
Abstract
Although antibody-drug conjugate (ADC) or immune checkpoint inhibitors (ICIs) alone fosters hope for the treatment of cancer, the effect of single drug treatment is limited and the safety profile of ADC and ICI therapy remains unclear. This meta-analysis aimed to examine the efficacy and safety of the combination of ADC and ICI therapy. This study type is a systematic review and meta-analysis. Literature retrieval was carried out through PubMed, Embase, Cochrane from inception to Jun. 5, 2024. Then, after data extraction, overall response rate (ORR) and adverse effects (AEs) were used to study its efficiency and safety. Publication bias was also calculated through Funnel plot, Begg's Test and Egger's test. Heterogeneity was investigated through subgroup and sensitivity analysis. The research protocol was registered with the PROSPERO (CRD42023375601). A total of 12 eligible clinical studies with 584 patients were included. The pooled ORR was 58% (95%CI 46%, 70%). Subgroup analysis showed an ORR of 77% (95%CI 63%, 91%) in classical Hodgkin lymphoma (cHL) and an ORR of 73% (95%CI 56%, 90%) in non-Hodgkin lymphoma (NHL). The most common AEs was peripheral neuropathy (38.0%). Meanwhile, AEs on skin (13.1-20.0%) and digestive system (9.0-36.0%) was hard be overlooked. ADC + ICI therapy may be recommended in cancer treatment, especially in cHL and NHL. However, strategies to manage toxicities warranted further exploration.
Collapse
Affiliation(s)
- Leyin Zhang
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yici Yan
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yangyang Gao
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yixin Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Jieru Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ning Ren
- Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China.
| | - Leitao Sun
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
5
|
Pan Y, Xue Q, Yang Y, Shi T, Wang H, Song X, Luo Y, Liu W, Ren S, Cai Y, Nie Y, Song Z, Liu B, Li JP, Wei J. Glycoengineering-based anti-PD-1-iRGD peptide conjugate boosts antitumor efficacy through T cell engagement. Cell Rep Med 2024; 5:101590. [PMID: 38843844 PMCID: PMC11228665 DOI: 10.1016/j.xcrm.2024.101590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024]
Abstract
Despite the important breakthroughs of immune checkpoint inhibitors in recent years, the objective response rates remain limited. Here, we synthesize programmed cell death protein-1 (PD-1) antibody-iRGD cyclic peptide conjugate (αPD-1-(iRGD)2) through glycoengineering methods. In addition to enhancing tissue penetration, αPD-1-(iRGD)2 simultaneously engages tumor cells and PD-1+ T cells via dual targeting, thus mediating tumor-specific T cell activation and proliferation with mild effects on non-specific T cells. In multiple syngeneic mouse models, αPD-1-(iRGD)2 effectively reduces tumor growth with satisfactory biosafety. Moreover, results of flow cytometry and single-cell RNA-seq reveal that αPD-1-(iRGD)2 remodels the tumor microenvironment and expands a population of "better effector" CD8+ tumor infiltrating T cells expressing stem- and memory-associated genes, including Tcf7, Il7r, Lef1, and Bach2. Conclusively, αPD-1-(iRGD)2 is a promising antibody conjugate therapeutic beyond antibody-drug conjugate for cancer immunotherapy.
Collapse
Affiliation(s)
- Yunfeng Pan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Qi Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Yi Yang
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Tao Shi
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hanbing Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xueru Song
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuting Luo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenqi Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shiji Ren
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiran Cai
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yang Nie
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhentao Song
- Glyco-therapy Biotechnology Co. Ltd., Building 12, Hangzhou Pharmaceutical Town, 291 Fucheng Road, Xiasha Street, Qiantang District, Hangzhou, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie P Li
- Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| | - Jia Wei
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China; Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Wang L, Jiang H, Yin X, Liang T, Li G, Ding C, Yang M, Zhang L, Liu J, Xu Y. PHE1-based IgG-like antibody platform provides a novel strategy for enhanced T-cell immunotherapy. Front Immunol 2024; 15:1415834. [PMID: 38933272 PMCID: PMC11201533 DOI: 10.3389/fimmu.2024.1415834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction Bispecific antibodies (BsAbs) can simultaneously target two epitopes of different antigenic targets, bringing possibilities for diversity in antibody drug design and are promising tools for the treatment of cancers and other diseases. T-cell engaging bsAb is an important application of the bispecific antibody, which could promote T cell-mediated tumor cell killing by targeting tumor-associated antigen (TAA) and CD3 at the same time. Methods This study comprised antibodies purification, Elisa assay for antigen binding, cytotoxicity assays, T cell activation by flow cytometry in vitro and xenogenic tumor model in vivo. Results We present a novel bsAb platform named PHE-Ig technique to promote cognate heavy chain (HC)-light chain (LC) pairing by replacing the CH1/CL regions of different monoclonal antibodies (mAbs) with the natural A and B chains of PHE1 fragment of Integrin β2 based on the knob-in-hole (KIH) technology. We had also verified that PHE-Ig technology can be effectively used as a platform to synthesize different desired bsAbs for T-cell immunotherapy. Especially, BCMA×CD3 PHE-Ig bsAbs exhibited robust anti-multiple myeloma (MM) activity in vitro and in vivo. Discussion Moreover, PHE1 domain was further shortened with D14G and R41S mutations, named PHE-S, and the PHE-S-based BCMA×CD3 bsAbs also showed anti BCMA+ tumor effect in vitro and in vivo, bringing more possibilities for the development and optimization of different bsAbs. To sum up, PHE1-based IgG-like antibody platform for bsAb construction provides a novel strategy for enhanced T-cell immunotherapy.
Collapse
Affiliation(s)
- Lingbin Wang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haojie Jiang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuying Yin
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoming Li
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Ding
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mina Yang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Synvida Biotechnology Co., Ltd, Shanghai, China
| | - Yanyan Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
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
|
Tanaka T, Suzuki H, Ohishi T, Kaneko MK, Kato Y. Antitumor activities against breast cancers by an afucosylated anti-HER2 monoclonal antibody H 2 Mab-77-mG 2a -f. Cancer Sci 2024; 115:298-309. [PMID: 37942574 PMCID: PMC10823288 DOI: 10.1111/cas.16008] [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: 08/25/2023] [Revised: 10/14/2023] [Accepted: 10/22/2023] [Indexed: 11/10/2023] Open
Abstract
Breast cancer patients with high levels of human epidermal growth factor receptor 2 (HER2) expression have worse clinical outcomes. Anti-HER2 monoclonal antibody (mAb) is the most important therapeutic modality for HER2-positive breast cancer. We previously immunized mice with the ectodomain of HER2 to create the anti-HER2 mAb, H2 Mab-77 (mouse IgG1 , kappa). This was then altered to produce H2 Mab-77-mG2a -f, an afucosylated mouse IgG2a . In the present work, we examined the reactivity of H2 Mab-77-mG2a -f and antitumor effects against breast cancers in vitro and in vivo. BT-474, an endogenously HER2-expressing breast cancer cell line, was identified by H2 Mab-77-mG2a -f with a strong binding affinity (a dissociation constant [KD ]: 5.0 × 10-9 M). H2 Mab-77-mG2a -f could stain HER2 of breast cancer tissues in immunohistochemistry and detect HER2 protein in Western blot analysis. Furthermore, H2 Mab-77-mG2a -f demonstrated strong antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) for BT-474 cells. MDA-MB-468, a HER2-negative breast cancer cell line, was unaffected by H2 Mab-77-mG2a -f. Additionally, in the BT-474-bearing tumor xenograft model, H2 Mab-77-mG2a -f substantially suppressed tumor development when compared with the control mouse IgG2a mAb. In contrast, the HER2-negative MDA-MB-468-bearing tumor xenograft model showed no response to H2 Mab-77-mG2a -f. These findings point to the possibility of H2 Mab-77-mG2a -f as a treatment regimen by showing that it has antitumor effects on HER2-positive breast tumors.
Collapse
Affiliation(s)
- Tomohiro Tanaka
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Hiroyuki Suzuki
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), NumazuMicrobial Chemistry Research FoundationShizuokaJapan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of OncologyMicrobial Chemistry Research FoundationTokyoJapan
| | - Mika K. Kaneko
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| | - Yukinari Kato
- Department of Molecular PharmacologyTohoku University Graduate School of MedicineSendaiMiyagiJapan
- Department of Antibody Drug DevelopmentTohoku University Graduate School of MedicineSendaiMiyagiJapan
| |
Collapse
|
9
|
Wang Z, Liu Y, Xu Y, Lu L, Zhu Z, Lv B, Fang X, Tang Y, Wang J, Cheng Y, Hu Y, Lou J, Wu P, Liu C, Liu Y, Zeng X, Xu Q. Anti-HER2 biparatopic antibody KJ015 has near-native structure, functional balanced high affinity, and synergistic efficacy with anti-PD-1 treatment in vivo. MAbs 2024; 16:2412881. [PMID: 39381966 PMCID: PMC11469434 DOI: 10.1080/19420862.2024.2412881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
Currently approved human epidermal growth factor receptor 2 (HER2)-targeted antibody therapies are largely derived from trastuzumab, including trastuzumab-chemotherapy combinations, fixed-dose trastuzumab-pertuzumab combinations, and trastuzumab antibody-drug conjugates. To expand the options, bispecific antibodies, which may better utilize the benefits of combination therapy, are being developed. Among them, biparatopic antibodies (bpAbs) have shown improved efficacy compared to monoclonal antibody (mAb) combinations in HER2-positive patients. BpAbs bind two independent epitopes on the same antigen, which allows fine-tuning of mechanisms of action, including enhancement of on-target specificity and induction of strong antigen clustering due to the unique binding mode. To fully utilize the potential of bpAbs for anti-HER2 drug development, it is crucial to consider formats that offer stability and high-yield production, along with a functional balance between the two epitopes. In this study, we rationally designed a bpAb, KJ015, that shares a common light chain with two Fab arms and exhibits functionally balanced high affinity for two HER2 non-overlapping epitopes. KJ015 demonstrated high-expression titers over 7 g/L and stable physicochemical properties at elevated concentrations, facilitating subcutaneous administration with hyaluronidase. Moreover, KJ015 maintained comparable antibody-dependent cytotoxicity, phagocytosis, and complement-dependent cytotoxicity with trastuzumab plus pertuzumab. It exhibited enhanced synergy when administered subcutaneously with hyaluronidase and anti-PD-1 mAb in a mouse tumor model, suggesting promising clinical prospects for this combination.
Collapse
Affiliation(s)
- Zheng Wang
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Yu Liu
- Department of Oncology, Shanghai Tenth People’s Hospital, Tongji University Cancer Center, Shanghai, China
| | - Yunxia Xu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Lin Lu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Zhen Zhu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Baojie Lv
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Xin Fang
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Yao Tang
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Jinhua Wang
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Yu Cheng
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Ying Hu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Junwen Lou
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Peican Wu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Chendan Liu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Yanjun Liu
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
| | - Xin Zeng
- Shanghai Bao Pharmaceuticals Co.Ltd, Baoshan, Shanghai, China
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, China
| | - Qing Xu
- Department of Oncology, Shanghai Tenth People’s Hospital, Tongji University Cancer Center, Shanghai, China
| |
Collapse
|
10
|
Mariottini D, Bracaglia S, Barbero L, Fuchs SW, Saal C, Moniot S, Knuehl C, Baranda L, Ranallo S, Ricci F. Bispecific Antibody Detection Using Antigen-Conjugated Synthetic Nucleic Acid Strands. ACS Sens 2023; 8:4014-4019. [PMID: 37856082 PMCID: PMC10683503 DOI: 10.1021/acssensors.3c01717] [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: 08/18/2023] [Accepted: 10/13/2023] [Indexed: 10/20/2023]
Abstract
We report here the development of two different sensing strategies based on the use of antigen-conjugated nucleic acid strands for the detection of a bispecific antibody against the tumor-related proteins Mucin1 and epidermal growth factor receptor. Both approaches work well in serum samples (nanomolar sensitivity), show high specificity against the two monospecific antibodies, and are rapid. The results presented here demonstrate the versatility of DNA-based platforms for the detection of bispecific antibodies and could represent a versatile alternative to other more reagent-intensive and time-consuming analytical approaches.
Collapse
Affiliation(s)
- Davide Mariottini
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Sara Bracaglia
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Luca Barbero
- RBM-Merck
(an affiliate of Merck KGaA), Via Ribes 1, 10010 Turin, Italy
| | | | - Christoph Saal
- Merck
KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany
| | | | | | - Lorena Baranda
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Simona Ranallo
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Francesco Ricci
- Department
of Chemical Science and Technologies, University
of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
11
|
Li HY, Chen YL, Deng XN, Li HH, Tan J, Liu GJ, Zheng YJ, Pei M, Peng KT, Yue LL, Chen XJ, Liu Y, Zhao YS, Wang CH. Bispecific antibody targeting both B7-H3 and PD-L1 exhibits superior antitumor activities. Acta Pharmacol Sin 2023; 44:2322-2330. [PMID: 37328649 PMCID: PMC10618207 DOI: 10.1038/s41401-023-01118-2] [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: 03/06/2023] [Accepted: 05/28/2023] [Indexed: 06/18/2023] Open
Abstract
Clinical application of PD-1 and PD-L1 monoclonal antibodies (mAbs) is hindered by their relatively low response rates and the occurrence of drug resistance. Co-expression of B7-H3 with PD-L1 has been found in various solid tumors, and combination therapies that target both PD-1/PD-L1 and B7-H3 pathways may provide additional therapeutic benefits. Up to today, however, no bispecific antibodies targeting both PD-1 and B7-H3 have reached the clinical development stage. In this study, we generated a stable B7-H3×PD-L1 bispecific antibody (BsAb) in IgG1-VHH format by coupling a humanized IgG1 mAb against PD-L1 with a humanized camelus variable domain of the heavy-chain of heavy-chain antibody (VHH) against human B7-H3. The BsAb exhibited favorable thermostability, efficient T cell activation, IFN-γ production, and antibody-dependent cell-mediated cytotoxicity (ADCC). In a PBMC humanized A375 xenogeneic tumor model, treatment with BsAb (10 mg/kg, i.p., twice a week for 6 weeks) showed enhanced antitumor activities compared to monotherapies and, to some degree, combination therapies. Our results suggest that targeting both PD-1 and B7-H3 with BsAbs increases their specificities to B7-H3 and PD-L1 double-positive tumors and induces a synergetic effect. We conclude that B7-H3×PD-L1 BsAb is favored over mAbs and possibly combination therapies in treating B7-H3 and PD-L1 double-positive tumors.
Collapse
Affiliation(s)
- Hua-Ying Li
- Shanghai Mabstone Biotechnologies, Ltd., Shanghai, 201203, China
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi-Li Chen
- Shanghai Mabstone Biotechnologies, Ltd., Shanghai, 201203, China.
| | - Xiang-Nan Deng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Huan-Huan Li
- Shanghai Mabstone Biotechnologies, Ltd., Shanghai, 201203, China
| | - Jie Tan
- Shanghai Mabstone Biotechnologies, Ltd., Shanghai, 201203, China
| | - Guo-Jian Liu
- Dartsbio Pharmaceuticals, Ltd., Zhongshan, 528400, China
| | - Yu-Juan Zheng
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Min Pei
- Shanghai Mabstone Biotechnologies, Ltd., Shanghai, 201203, China
- Institute of Biomedicine & Department of Cell Biology, Jinan University, National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Kai-Ting Peng
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li-Li Yue
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao-Jia Chen
- Institute of Biomedicine & Department of Cell Biology, Jinan University, National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China
| | - Yu Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yong-Shan Zhao
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Chun-He Wang
- Biotherapeutics Discovery Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Dartsbio Pharmaceuticals, Ltd., Zhongshan, 528400, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Biomedicine & Department of Cell Biology, Jinan University, National Engineering Research Center of Genetic Medicine, Guangzhou, 510632, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
12
|
Pous A, Notario L, Hierro C, Layos L, Bugés C. HER2-Positive Gastric Cancer: The Role of Immunotherapy and Novel Therapeutic Strategies. Int J Mol Sci 2023; 24:11403. [PMID: 37511163 PMCID: PMC10380453 DOI: 10.3390/ijms241411403] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Gastric cancer is an aggressive disease with increasing global incidence in recent years. Human epidermal growth receptor 2 (HER2) is overexpressed in approximately 10-20% of gastric cancers. The implementation of targeted therapy against HER2 as part of the standard of care treatment in metastatic disease has improved the prognosis of this subset of patients. However, gastric cancer still has high mortality rates and urgently requires new treatment strategies. The combination of immunotherapy with HER2-targeted therapies has shown synergistic effects in preclinical models, this being the rationale behind exploring this combination in clinical trials in locally advanced and metastatic settings. Additionally, the irruption of antibody-drug conjugates and other novel HER2-targeted agents has led to the development of numerous clinical trials showing promising results. This review presents the molecular mechanisms supporting the use of HER2-targeted drugs in combination with immunotherapy and provides an overview of the therapeutic scenario of HER2-positive disease. We focus on the role of immunotherapy but also summarize emerging therapies and combinations under clinical research that may change the standard treatment in HER-2 positive disease in the future.
Collapse
Affiliation(s)
- Anna Pous
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Lucía Notario
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Cinta Hierro
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Laura Layos
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| | - Cristina Bugés
- Department of Medical Oncology, Institut Català d'Oncologia (ICO) Badalona, 08916 Badalona, Spain
- Badalona Applied Research Group in Oncology (B-ARGO), 08916 Badalona, Spain
| |
Collapse
|
13
|
Burgess EF, Livasy CA, Symanowski JT, Matulay JT, Grigg CM, Clark PE, Raghavan D. Low co-expression of PD-L1 and oncogenic receptor tyrosine kinases HER2 and cMET in urothelial carcinoma is associated with discordant expression between primary and metastatic sites. Urol Oncol 2023:S1078-1439(23)00187-4. [PMID: 37295980 DOI: 10.1016/j.urolonc.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVES Novel regimens targeting immune checkpoints and the cMET or HER2 pathways are under investigation in metastatic urothelial carcinoma (mUC) though co-expression of these molecular targets has not been defined. We sought to characterize the protein co-expression rates of PD-L1, cMET and HER2 in primary and metastatic mUC lesions and agreement rates in paired biopsies. MATERIALS AND METHODS We assessed PD-L1, cMET and HER2 protein expression by immunohistochemistry (IHC) in archival mUC samples identified from an institutional database (n = 143). Correlation of expression between primary and metastatic biopsies was performed in patients with available paired biopsies (n = 79). Protein expression levels by predefined thresholds were measured, and Cohen's kappa statistics (κ) were utilized to assess the agreement in expression between paired primary and metastatic samples. RESULTS In primary tumors (n = 85), high expression of PD-L1, cMET, and HER2 was observed in 14.1%, 34.1%, and 12.9%, respectively. In metastatic samples (n = 143), high expression of PD-L1, cMET and HER2 was detected in 9.8%, 41.3%, and 9.8%, respectively. Expression agreement rates between paired specimens (n = 79) were PD-L1: 79.7% (κ = 0.09), cMET: 69.6% (κ = 0.35), HER2: 84.8% (κ = 0.17). High PD-L1/cMET co-expression was observed in only 5.1% (n = 4) of primary and 4.9% (n = 7) of metastatic specimens. High co-expression of PD-L1/HER2 occurred in 3.8% (n = 3) of primary samples and no metastatic samples. The overall co-expression agreement between paired samples was 55.7% (κ = 0.22) for PD-L1/cMET and 67.1% (κ = 0.06) for PD-L1/HER2, but agreement for high co-expression between paired samples was very low (2.5% for PD-L1/cMET and 0% for PD-L1/HER2). CONCLUSIONS Tumor co-expression of high cMET or HER2 and PD-L1 is low in this cohort. Agreement of high co-expression between primary and metastatic sites is rare. Biomarker-based strategies used in selection of patients for contemporary trials testing combinations of immune checkpoint inhibitors with either cMET or HER2-targeted agents should account for discordant biomarker expression between primary and metastatic sites.
Collapse
Affiliation(s)
- Earle F Burgess
- Department of Solid Tumor Oncology, Atrium Health, Levine Cancer Institute; Charlotte, NC.
| | - Chad A Livasy
- Department of Pathology, Atrium Health, Levine Cancer Institute, Charlotte, NC
| | - James T Symanowski
- Department of Biostatistics and Data Sciences, Atrium Health, Levine Cancer Institute, Charlotte, NC
| | | | - Claud M Grigg
- Department of Solid Tumor Oncology, Atrium Health, Levine Cancer Institute; Charlotte, NC
| | | | - Derek Raghavan
- Department of Solid Tumor Oncology, Atrium Health, Levine Cancer Institute; Charlotte, NC
| |
Collapse
|
14
|
Mortezaee K, Majidpoor J. Reinstating immunogenicity using bispecific anti-checkpoint/agent inhibitors. Biomed Pharmacother 2023; 162:114621. [PMID: 37004328 DOI: 10.1016/j.biopha.2023.114621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Immune checkpoint inhibitor (ICI) resistance demands for acquisition of novel strategies in order to broaden the therapeutic repertoire of advanced cancers. Bispecific antibodies can be utilized as an emerging therapeutic paradigm and a step forward in cancer immunotherapy. Synchronous inhibition of programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1) or cytotoxic T lymphocyte associated antigen-4 (CTLA-4), or with other agents can expand antibody selectivity and improve therapeutic window through tightening cell-to-cell bridge (a process called immunological synapse) within tumor immune microenvironment (TIME). There is evidence of higher potency of this co-targeting approach over combined single-agent monoclonal antibodies in reinvigorating anti-tumor immune responses, retarding tumor growth, and improving patient survival. In fact, immunological synapses formed by interactions of such bispecific agents with TIME cells directly mediate cytotoxicity against tumor cells, and durable anti-tumor immune responses are predictable after application of such agents. Besides, lower adverse events are reported for bispecific antibodies compared with individual checkpoint inhibitors. These are all indicative of the importance of exploiting novel bispecific approach as a replacement for conventional combo checkpoint inhibitor therapy particularly for tumors with immunosuppressive or cold immunity. Study in this area is still continued, and in the future more will be known about the importance of this bispecific approach in cancer immunotherapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
15
|
Jacquot P, Muñoz-Garcia J, Fleury M, Cochonneau D, Gaussin R, Enouf E, Roze C, Ollivier E, Cinier M, Heymann D. Engineering of a Bispecific Nanofitin with Immune Checkpoint Inhibitory Activity Conditioned by the Cross-Arm Binding to EGFR and PDL1. Biomolecules 2023; 13:biom13040636. [PMID: 37189383 DOI: 10.3390/biom13040636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Re-education of the tumor microenvironment with immune checkpoint inhibitors (ICI) has provided the most significant advancement in cancer management, with impressive efficacy and durable response reported. However, low response rates and a high frequency of immune-related adverse events (irAEs) remain associated with ICI therapies. The latter can be linked to their high affinity and avidity for their target that fosters on-target/off-tumor binding and subsequent breaking of immune self-tolerance in normal tissues. Many multispecific protein formats have been proposed to increase the tumor cell’s selectivity of ICI therapies. In this study, we explored the engineering of a bispecific Nanofitin by the fusion of an anti-epidermal growth factor receptor (EGFR) and anti-programmed cell death ligand 1 (PDL1) Nanofitin modules. While lowering the affinity of the Nanofitin modules for their respective target, the fusion enables the simultaneous engagement of EGFR and PDL1, which translates into a selective binding to tumor cells co-expressing EGFR and PDL1 only. We demonstrated that affinity-attenuated bispecific Nanofitin could elicit PDL1 blockade exclusively in an EGFR-directed manner. Overall, the data collected highlight the potential of this approach to enhance the selectivity and safety of PDL1 checkpoint inhibition.
Collapse
|
16
|
Mercogliano MF, Bruni S, Mauro FL, Schillaci R. Emerging Targeted Therapies for HER2-Positive Breast Cancer. Cancers (Basel) 2023; 15:cancers15071987. [PMID: 37046648 PMCID: PMC10093019 DOI: 10.3390/cancers15071987] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Breast cancer is the most common cancer in women and the leading cause of death. HER2 overexpression is found in approximately 20% of breast cancers and is associated with a poor prognosis and a shorter overall survival. Tratuzumab, a monoclonal antibody directed against the HER2 receptor, is the standard of care treatment. However, a third of the patients do not respond to therapy. Given the high rate of resistance, other HER2-targeted strategies have been developed, including monoclonal antibodies such as pertuzumab and margetuximab, trastuzumab-based antibody drug conjugates such as trastuzumab-emtansine (T-DM1) and trastuzumab-deruxtecan (T-DXd), and tyrosine kinase inhibitors like lapatinib and tucatinib, among others. Moreover, T-DXd has proven to be of use in the HER2-low subtype, which suggests that other HER2-targeted therapies could be successful in this recently defined new breast cancer subclassification. When patients progress to multiple strategies, there are several HER2-targeted therapies available; however, treatment options are limited, and the potential combination with other drugs, immune checkpoint inhibitors, CAR-T cells, CAR-NK, CAR-M, and vaccines is an interesting and appealing field that is still in development. In this review, we will discuss the highlights and pitfalls of the different HER2-targeted therapies and potential combinations to overcome metastatic disease and resistance to therapy.
Collapse
|
17
|
Guo L, Overholser J, Darby H, Ede NJ, Kaumaya PT. A newly discovered PD-L1 B-cell epitope peptide vaccine (PDL1-Vaxx) exhibits potent immune responses and effective anti-tumor immunity in multiple syngeneic mice models and (synergizes) in combination with a dual HER-2 B-cell vaccine (B-Vaxx). Oncoimmunology 2022; 11:2127691. [PMID: 36211807 PMCID: PMC9542669 DOI: 10.1080/2162402x.2022.2127691] [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: 11/09/2022] Open
Abstract
Blockade of checkpoint receptors with monoclonal antibodies against CTLA-4, PD-1 and PD-L1 has shown great clinical success in several cancer subtypes, yielding unprecedented responses albeit a significant number of patients develop resistance and remain refractory. Both PD-1/PD-L1 and HER-2 signaling pathway inhibitors have limited efficacy and exhibits significant toxicities that limit their use. Ongoing clinical studies support the need for rationale combination of immuno-oncology agents to make a significant impact in the lives of cancer patients. We introduce the development of a novel chimeric PD-L1 B-cell peptide epitope vaccine (amino acid 130–147) linked to a “promiscuous” T cell measles virus fusion (MVF) peptide (MVF-PD-L1(130); PDL1-Vaxx) or linked to tetanus toxoid (TT3) TT3-PD-L1 (130) via a linker (GPSL). These vaccine constructs are highly immunogenic and antigenic in several syngeneic animal models. The PD-L1 vaccines elicited high titers of polyclonal antibodies that inhibit tumor growth in multiple syngeneic cancer models, eliciting antibodies of different subtypes IgG1, IgG2a, IgG2b and IgG3, induced PD-1/PD-L1 blockade, decreased proliferation, induced apoptosis and caused ADCC of tumor cells. The PDL1-Vaxx induces similar inhibition of tumor growth versus the standard anti-mouse PD-L1 antibody in both syngeneic BALB/c and C57BL/6J mouse models. The combination of PDL1-Vaxx with HER-2 vaccine B-Vaxx demonstrated synergistic tumor inhibition in D2F2/E2 carcinoma cell line. The anti-PDL1-Vaxx block PD-1/PD-L1 interaction and significantly prolonged anti-tumor responses in multiple syngeneic tumor models. The combination of HER-2 vaccine (B-Vaxx) with either PDL1-Vaxx or PD1-Vaxx demonstrated synergistic tumor inhibition. PDL1-Vaxx is a promising novel safe checkpoint inhibitor vaccine.
Collapse
Affiliation(s)
- Linlin Guo
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, USA
| | - Jay Overholser
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, USA
| | - Heather Darby
- Licensing Technology, Luminex Corp, Austin Texas, USA
| | | | - Pravin T.P Kaumaya
- Department of Obstetrics & Gynecology, The Ohio State University Wexner Medical Center, USA
- The James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
18
|
Aisa A, Weng S, Li X, Zhang D, Yuan Y. Immune Checkpoint Inhibitors combined with HER-2 targeted therapy in HER-2 positive Gastroesophageal cancer. Crit Rev Oncol Hematol 2022; 180:103864. [DOI: 10.1016/j.critrevonc.2022.103864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
|
19
|
Chow CY, Lie EF, Wu CH, Chow LW. Clinical implication of genetic composition and molecular mechanism on treatment strategies of HER2-positive breast cancers. Front Oncol 2022; 12:964824. [PMID: 36387174 PMCID: PMC9659858 DOI: 10.3389/fonc.2022.964824] [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] [Received: 06/09/2022] [Accepted: 10/14/2022] [Indexed: 12/01/2022] Open
Abstract
The current clinical management model of HER2-positive breast cancers is commonly based on guidelines, which in turn are based on the design and outcome of clinical trials. While this model is useful to most practicing clinicians, the treatment outcome of individual patient is not certain at the start of treatment. As the understanding of the translational research of carcinogenesis and the related changes in cancer genetics and tumor microenvironment during treatment is critical in the selection of right choice of treatment to maximize the successful clinical outcome for the patient, this review article intends to discuss the latest developments in the genetic and molecular mechanisms of cancer progression and treatment resistance, and how they influence the planning of the treatment strategies of HER2-positive breast cancers.
Collapse
Affiliation(s)
- Christopher Y.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | | | - Cheng-Hsun Wu
- Department of Anatomy, China Medical University, Taichung, Taiwan
| | - Louis W.C. Chow
- UNIMED Medical Institute, Hong Kong, Hong Kong SAR, China
- Organisation for Oncology and Translational Research, Hong Kong, Hong Kong SAR, China
- *Correspondence: Louis W.C. Chow,
| |
Collapse
|
20
|
Pei M, Wang Y, Tang L, Wu W, Wang C, Chen YL. Dual-target Bridging ELISA for Bispecific Antibodies. Bio Protoc 2022; 12:e4522. [PMID: 36313202 PMCID: PMC9548515 DOI: 10.21769/bioprotoc.4522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/20/2022] [Accepted: 09/05/2022] [Indexed: 12/29/2022] Open
Abstract
Bispecific antibodies (BsAbs) are typically monoclonal antibody (mAb)-derived molecular entities engineered to bind to two distinct targets, including two antigens or two epitopes on the same antigen. When compared to parental monoclonal antibodies or combinational therapies, the generated BsAbs have the ability to bridge the two targets and thus may offer additional clinical benefits. Characterizing BsAbs' ability to bind to both targets simultaneously is critical for their biotherapeutic development. A range of bi-functional quantitative bridging assays to enable target-specific capture and detection of binding properties include enzyme-linked immunosorbent assay (ELISA), surface plasmon resonance (SPR), and cell-based flow cytometry. Developing suitable and robust cell-based bioassays is more challenging than non-cell-based binding assays because cell-based assays with complex matrices can be inherently variable and often lack precision. Compared to SPR, ELISA has a rapid setup and readily available method, being widely and extensively applied in almost every laboratory. Here, we describe a dual-target bridging ELISA assay that characterizes the ability of a HER2(human epidermal growth factor receptor 2)/PD-L1(programmed cell death ligand 1) BsAb in binding to both HER2 and PD-L1 simultaneously, a prerequisite for its envisioned mode of action. Graphical abstract.
Collapse
Affiliation(s)
- Min Pei
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
| | - Yao Wang
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
| | - Lei Tang
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
| | - Weitao Wu
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
| | - Chunhe Wang
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
,
Department of Research and Development Center, Dartsbio Pharmaceuticals Ltd, Zhongshan, Guangdong, China
,
*For correspondence:
;
| | - Yi-Li Chen
- Department of Antibody Discovery, Shanghai Mabstone Biotechnology, Ltd, Shanghai, China
,
*For correspondence:
;
| |
Collapse
|
21
|
Chen W, Huang Y, Pan W, Xu M, Chen L. Strategies for developing PD-1 inhibitors and future directions. Biochem Pharmacol 2022; 202:115113. [DOI: 10.1016/j.bcp.2022.115113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023]
|