1
|
Yi L, Xu Z, Ma T, Wang C, Wei P, Xiao B, Zhang H, Che N, Liu Z, Han Y. T-cell subsets and cytokines are indicative of neoadjuvant chemoimmunotherapy responses in NSCLC. Cancer Immunol Immunother 2024; 73:99. [PMID: 38619623 PMCID: PMC11018727 DOI: 10.1007/s00262-024-03687-5] [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: 08/24/2023] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE Neoadjuvant PD-1 blockade combined with chemotherapy is a promising treatment for resectable non-small cell lung cancer (NSCLC), yet the immunological mechanisms contributing to tumor regression and biomarkers corresponding to different pathological responses remain unclear. METHODS Using dynamic and paired blood samples from NSCLC patients receiving neoadjuvant chemoimmunotherapy, we analyzed the frequencies of CD8 + T-cell and Treg subsets and their dynamic changes during neoadjuvant treatment through flow cytometry. Cytokine profiles and function-related gene expression of CD8 + T cells and Tregs were analyzed through flow cytometry and mRNA-seq. Infiltrating T-cell subsets in resected tissues from patients with different pathological responses were analyzed through multiplex immunofluorescence. RESULTS Forty-two NSCLC patients receiving neoadjuvant chemoimmunotherapy were enrolled and then underwent surgical resection and pathological evaluation. Nineteen patients had pCR (45%), 7 patients had MPR (17%), and 16 patients had non-MPR (38%). In patients with pCR, the frequencies of CD137 + CD8 + T cells (P = 0.0475), PD-1 + Ki-67 + CD8 + T cells (P = 0.0261) and Tregs (P = 0.0317) were significantly different from those of non-pCR patients before treatment. pCR patients usually had low frequencies of CD137 + CD8 + T cells, PD-1 + Ki-67 + CD8 + T cells and Tregs, and their AUCs were higher than that of tissue PD-L1 expression. Neoadjuvant chemoimmunotherapy markedly improved CD8 + T-cell proliferation and activation, especially in pCR patients, as the frequencies of CD137 + CD8 + (P = 0.0136) and Ki-67 + CD8 + (P = 0.0391) T cells were significantly increased. The blood levels of cytokines such as IL-2 (P = 0.0391) and CXCL10 (P = 0.0195) were also significantly increased in the pCR group, which is consistent with the high density of activated cytotoxic T cells at the tumor site (P < 0.0001). CONCLUSION Neoadjuvant chemoimmunotherapy drives CD8 + T cells toward a proliferative and active profile. The frequencies of CD137 + CD8 + T cells, PD-1 + Ki-67 + CD8 + T cells and Tregs at baseline might predict the response to neoadjuvant chemoimmunotherapy in NSCLC patients. The increase in IL-2 and CXCL10 might reflect the chemotaxis and enrichment of cytotoxic T cells at the tumor site and a better response to neoadjuvant chemoimmunotherapy.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Ziwei Xu
- Department of Minimally Invasive Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Tianyu Ma
- Department of Thoracic Surgery II, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Chong Wang
- Department of Minimally Invasive Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Panjian Wei
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bo Xiao
- Department of Minimally Invasive Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| | - Nanying Che
- Department of Pathology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| | - Zhidong Liu
- Department of Thoracic Surgery II, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| | - Yi Han
- Department of Minimally Invasive Surgery, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Na K, Lee S, Kim DK, Kim YS, Hwang JY, Kang SS, Baek S, Lee CY, Yang SM, Han YJ, Kim MH, Han H, Kim Y, Kim JH, Jeon S, Byeon Y, Lee JB, Lim SM, Hong MH, Pyo KH, Cho BC. CD81 and CD82 expressing tumor-infiltrating lymphocytes in the NSCLC tumor microenvironment play a crucial role in T-cell activation and cytokine production. Front Immunol 2024; 15:1336246. [PMID: 38515751 PMCID: PMC10954780 DOI: 10.3389/fimmu.2024.1336246] [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: 11/10/2023] [Accepted: 02/02/2024] [Indexed: 03/23/2024] Open
Abstract
Introduction To understand the immune system within the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC), it is crucial to elucidate the characteristics of molecules associated with T cell activation. Methods We conducted an in-depth analysis using single-cell RNA sequencing data obtained from tissue samples of 19 NSCLC patients. T cells were classified based on the Tumor Proportion Score (TPS) within the tumor region, and molecular markers associated with activation and exhaustion were analyzed in T cells from high TPS areas. Results Notably, tetraspanins CD81 and CD82, belonging to the tetraspanin protein family, were found to be expressed in activated T cells, particularly in cytotoxic T cells. These tetraspanins showed strong correlations with activation and exhaustion markers. In vitro experiments confirmed increased expression of CD81 and CD82 in IL-2-stimulated T cells. T cells were categorized into CD81highCD82high and CD81lowCD82low groups based on their expression levels, with CD81highCD82high T cells exhibiting elevated activation markers such as CD25 and CD69 compared to CD81lowCD82low T cells. This trend was consistent across CD3+, CD8+, and CD4+ T cell subsets. Moreover, CD81highCD82high T cells, when stimulated with anti-CD3, demonstrated enhanced secretion of cytokines such as IFN-γ, TNF-α, and IL-2, along with an increase in the proportion of memory T cells. Bulk RNA sequencing results after sorting CD81highCD82high and CD81lowCD82low T cells consistently supported the roles of CD81 and CD82. Experiments with overexpressed CD81 and CD82 showed increased cytotoxicity against target cells. Discussion These findings highlight the multifaceted roles of CD81 and CD82 in T cell activation, cytokine production, memory subset accumulation, and target cell cytolysis. Therefore, these findings suggest the potential of CD81 and CD82 as promising candidates for co-stimulatory molecules in immune therapeutic strategies for cancer treatment within the intricate TME.
Collapse
Affiliation(s)
- Kwangmin Na
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seul Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Dong Kwon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | - Young Seob Kim
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Yeon Hwang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seong-San Kang
- JEUK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Republic of Korea
| | - Sujeong Baek
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chai Young Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Min Yang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Jin Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi Hyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heekyung Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngtaek Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seunghyun Jeon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngseon Byeon
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Min Lim
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Ho Pyo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoung Chul Cho
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
4
|
Kim JH, Kim GH, Ryu YM, Kim SY, Kim HD, Yoon SK, Cho Y, Lee JL. Clinical implications of the tumor microenvironment using multiplexed immunohistochemistry in patients with advanced or metastatic renal cell carcinoma treated with nivolumab plus ipilimumab. Front Oncol 2022; 12:969569. [PMID: 36237314 PMCID: PMC9552830 DOI: 10.3389/fonc.2022.969569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/13/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Immune checkpoint inhibitors (ICIs) such as nivolumab and ipilimumab (N/I) are important treatment options for advanced renal cell carcinoma (RCC). The tumor microenvironment (TME) in these ICI-treated patients is largely unknown. Methods Twenty-four patients treated with N/I between July 2015 and June 2020 were analyzed. Multiplexed immunohistochemistry (mIHC) was conducted to define the TME, including various T cell subsets, B cells, macrophages, and dendritic cells. Results The median age of the study patients was 61 years (range, 39-80) and 75.0% of these cases were men. The objective response rate with N/I was 50.0%. The densities of the CD8+ cytotoxic T cells (P=0.005), specifically CD137+ CD8+ T cells (P=0.017), Foxp3- CD4+ helper T cells (P=0.003), Foxp3+ CD4+ regulatory T cells (P=0.045), CD68+ CD206- M1 macrophages (P=0.008), and CD68+ CD206+ M2 macrophages (P=0.021) were significantly higher in the treatment responders. At a median follow-up duration of 24.7 months, the median progression-free survival (PFS) was 11.6 months. The high densities (≥median) of Foxp3- CD4+ helper T cells (P=0.016) and CD68+ CD206- M1 macrophages (P=0.008) were significantly associated with better PFS, and the density of CD137+ CD8+ cytotoxic T cells (P=0.079) was marginally associated with better PFS. After multivariate analysis, the higher density of Foxp3- CD4+ helper T cells was independently associated with better PFS (hazard ratio 0.19; P=0.016). Conclusion The properties and clinical implications of the TME properties in RCC indicate that Foxp3- CD4+ helper T cells, M1 macrophages, and CD137+ CD8+ T cells are potential predictive biomarkers and treatment targets.
Collapse
Affiliation(s)
- Jwa Hoon Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Gi Hwan Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yeon-Mi Ryu
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Sang-Yeob Kim
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Shin Kyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong Mee Cho
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
5
|
Immune-associated plasma proteins in oral and oropharyngeal cancer patients. Heliyon 2022; 8:e10753. [PMID: 36193525 PMCID: PMC9525905 DOI: 10.1016/j.heliyon.2022.e10753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/21/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Background Plasma protein patterns differ between cancer patients and healthy donors. This study aimed to examine the plasma levels of several cytokines and immunological checkpoint proteins between patients with oral and oropharyngeal cancer and healthy donors. Materials and methods Plasma samples from healthy donors, oral cancer patients, and oropharyngeal cancer patients were analyzed using the Human Th Cytokine Panel 13-plex (IL-2, 4, 5, 6, 9, 10, 13, 17A, 17F, 21, 22, IFN-γ, and TNF-α) and Human Immune Checkpoint Panel1 12-plex [sCD25 (IL-2Ra), 4-1BB, sCD27, B7.2 (CD86), Free Active TGF-β1, CTLA-4, PD-L1, PD-L2, PD-1, Tim-3, LAG-3, and Galectin-9]. The plasma 4-1BB levels were verified by Western blot method. In addition, the study of the receive operating curve (ROC) yielded the calculation of a number of diagnostically significant indicators. Results Significantly increased levels of IL-6, 4-1BB, PDL-1, PD-1, and CTLA-4 and decreased levels of IL-13 and sCD27 were observed in cancer patients compared with healthy donors. These levels were highly significant, particularly for cancer patients in stage IV. Validation by Western blot revealed that cancer patients had higher plasma levels of 4-1BB than healthy donors (p < 0.05), and ROC curve analysis revealed that plasma 4-1BB had the highest cancer detection capability. Intriguingly, plasma levels of 4-1BB were significantly positively correlated with PDL-1 and PD-1 levels (p < 0.0001). Conclusion This data provided descriptive knowledge of oral and oropharyngeal cancer immunity at a fundamental level. Additional research should concentrate on the significantly different factors, especially 4-1BB, PDL-1, and PD-1, which may contribute to the development of novel alternative diagnostic tools or therapies for patients with oral and oropharyngeal cancer. Plasma levels of 4-1BB were increased in head and neck cancer patients. The level of plasma 4-1BB correlated with levels of plasma PD-1 and PDL-1. Plasma 4-1BB possible to use as head and neck cancer screening. Plasma 4-1BB may be developed as an alternative cancer immunotherapeutic drug.
Collapse
|
6
|
Hussain Y, Mirzaei S, Ashrafizadeh M, Zarrabi A, Hushmandi K, Khan H, Daglia M. Quercetin and Its Nano-Scale Delivery Systems in Prostate Cancer Therapy: Paving the Way for Cancer Elimination and Reversing Chemoresistance. Cancers (Basel) 2021; 13:1602. [PMID: 33807174 PMCID: PMC8036441 DOI: 10.3390/cancers13071602] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 03/04/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is the second most leading and prevalent malignancy around the world, following lung cancer. Prostate cancer is characterized by the uncontrolled growth of cells in the prostate gland. Prostate cancer morbidity and mortality have grown drastically, and intensive prostate cancer care is unlikely to produce adequate outcomes. The synthetic drugs for the treatment of prostate cancer in clinical practice face several challenges. Quercetin is a natural flavonoid found in fruits and vegetables. Apart from its beneficial effects, its plays a key role as an anti-cancer agent. Quercetin has shown anticancer potential, both alone and in combination. Therefore, the current study was designed to collect information from the literature regarding its therapeutic significance in the treatment of prostate cancer. Studies performed both in vitro and in vivo have confirmed that quercetin effectively prevents prostate cancer through different underlying mechanisms. Promising findings have also been achieved in clinical trials regarding the pharmacokinetics and human applications of quercetin. In the meantime, epidemiological studies have shown a negative correlation between the consumption of quercetin and the incidence of prostate cancer, and have indicated a chemopreventive effect of quercetin on prostate cancer in animal models. The major issues associated with quercetin are its low bioavailability and rapid metabolism, and these require priority attention. Chemoresistance is another main negative feature concerning prostate cancer treatment. This review highlights the chemotherapeutic effect, chemo preventive effect, and chemoresistance elimination potential of quercetin in prostate cancer. The underlying mechanisms for elimination of prostate cancer and eradication of resistance, either alone or in combination with other agents, are also discussed. In addition, the nanoscale delivery of quercetin is underpinned along with possible directions for future study.
Collapse
Affiliation(s)
- Yaseen Hussain
- Lab of Control Release and Drug Delivery System, College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China;
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran 1477893855, Iran;
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul 34956, Turkey;
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417466191, Iran;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, 80131 Naples, Italy;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
7
|
Kuriyama H, Fukushima S, Kimura T, Kanemaru H, Miyashita A, Okada E, Kubo Y, Nakahara S, Tokuzumi A, Nishimura Y, Kajihara I, Makino K, Aoi J, Masuguchi S, Tsukamoto H, Inozume T, Zhang R, Nakatsura T, Uemura Y, Senju S, Ihn H. Immunotherapy with 4-1BBL-Expressing iPS Cell-Derived Myeloid Lines Amplifies Antigen-Specific T Cell Infiltration in Advanced Melanoma. Int J Mol Sci 2021; 22:1958. [PMID: 33669419 PMCID: PMC7920470 DOI: 10.3390/ijms22041958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 11/16/2022] Open
Abstract
We have established an immune cell therapy with immortalized induced pluripotent stem-cell-derived myeloid lines (iPS-ML). The benefits of using iPS-ML are the infinite proliferative capacity and ease of genetic modification. In this study, we introduced 4-1BBL gene to iPS-ML (iPS-ML-41BBL). The analysis of the cell-surface molecules showed that the expression of CD86 was upregulated in iPS-ML-41BBL more than that in control iPS-ML. Cytokine array analysis was performed using supernatants of the spleen cells that were cocultured with iPS-ML or iPS-ML-41BBL. Multiple cytokines that are beneficial to cancer immunotherapy were upregulated. Peritoneal injections of iPS-ML-41BBL inhibited tumor growth of peritoneally disseminated mouse melanoma and prolonged survival of mice compared to that of iPS-ML. Furthermore, the numbers of antigen-specific CD8+ T cells were significantly increased in the spleen and tumor tissues treated with epitope peptide-pulsed iPS-ML-41BBL compared to those treated with control iPS-ML. The number of CXCR6-positive T cells were increased in the tumor tissues after treatment with iPS-ML-41BBL compared to that with control iPS-ML. These results suggest that iPS-ML-41BBL could activate antigen-specific T cells and promote their infiltration into the tumor tissues. Thus, iPS-ML-41BBL may be a candidate for future immune cell therapy aiming to change immunological "cold tumor" to "hot tumor".
Collapse
Affiliation(s)
- Haruka Kuriyama
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Toshihiro Kimura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Hisashi Kanemaru
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Etsuko Okada
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Yosuke Kubo
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Satoshi Nakahara
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Aki Tokuzumi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Yuki Nishimura
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Ikko Kajihara
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Katsunari Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Jun Aoi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Shinichi Masuguchi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| | - Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| | - Takashi Inozume
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan;
| | - Rong Zhang
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center (NCC), Kashiwa 277-8577, Japan; (R.Z.); (T.N.); (Y.U.)
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center (NCC), Kashiwa 277-8577, Japan; (R.Z.); (T.N.); (Y.U.)
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center (NCC), Kashiwa 277-8577, Japan; (R.Z.); (T.N.); (Y.U.)
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan;
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan; (H.K.); (T.K.); (H.K.); (A.M.); (E.O.); (Y.K.); (S.N.); (A.T.); (Y.N.); (I.K.); (K.M.); (J.A.); (S.M.); (H.I.)
| |
Collapse
|
8
|
Fucà G, Spagnoletti A, Ambrosini M, de Braud F, Di Nicola M. Immune cell engagers in solid tumors: promises and challenges of the next generation immunotherapy. ESMO Open 2021; 6:100046. [PMID: 33508733 PMCID: PMC7841318 DOI: 10.1016/j.esmoop.2020.100046] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 12/22/2022] Open
Abstract
In the landscape of cancer immunotherapy, immune cell engagers (ICEs) are rapidly emerging as a feasible and easy-to-deliver alternative to adoptive cell therapy for the antitumor redirection of immune effector cells. Even if in hematological malignancies this class of new therapeutics already hit the clinic, the development of ICEs in solid tumors still represents a challenge. Considering that ICEs are a rapidly expanding biotechnology in cancer therapy, we designed this review as a primer for clinicians, focusing on the major obstacles for the clinical implementation and the most translatable approaches proposed to overcome the limitations in solid tumors.
Collapse
Affiliation(s)
- G Fucà
- Immunotherapy and Innovative Therapeutics Unit, Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - A Spagnoletti
- Immunotherapy and Innovative Therapeutics Unit, Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Ambrosini
- Immunotherapy and Innovative Therapeutics Unit, Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F de Braud
- Immunotherapy and Innovative Therapeutics Unit, Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, Milan, Italy
| | - M Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Medical Oncology and Hematology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
9
|
Kwon M, Jung H, Nam GH, Kim IS. The right Timing, right combination, right sequence, and right delivery for Cancer immunotherapy. J Control Release 2021; 331:321-334. [PMID: 33434599 DOI: 10.1016/j.jconrel.2021.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy (CI) represented by immune checkpoint inhibitors (ICIs) presents a new paradigm for cancer treatment. However, the types of cancer that attain a therapeutic benefit from ICIs are limited, and the efficacy of these treatments does not meet expectations. To date, research on ICIs has mainly focused on identifying biomarkers and patient characteristics that can enhance the therapeutic effect on tumors. However, studies on combinational strategies for CI are being actively conducted to overcome the resistance to ICI treatment. Moreover, it has been confirmed that dramatic anticancer effects are achieved through "neoadjuvant" immunotherapy with ICIs in treatment-naïve cancer patients; consequently, it has become necessary to consider how to best apply cancer immunotherapies for patients, even with respect to their tumor stages. In this review, we sought to discuss the right timing of ICI treatment in consideration of the progression of cancer with a changing tumor-immune microenvironment. Furthermore, we investigated which types of combinational treatments and their corresponding sequences of administration could optimize the therapeutic effect of ICIs to expand the applicable target of ICIs and increase their therapeutic efficacy. Finally, we discussed several delivery pathways and methods that can maximize the effect of ICIs.
Collapse
Affiliation(s)
- Minsu Kwon
- Korea University Anam Hospital, Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea.
| | - Hanul Jung
- Korea University Anam Hospital, Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, Republic of Korea
| | - Gi-Hoon Nam
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, Republic of Korea.
| |
Collapse
|
10
|
Zizzari IG, Napoletano C, Di Filippo A, Botticelli A, Gelibter A, Calabrò F, Rossi E, Schinzari G, Urbano F, Pomati G, Scagnoli S, Rughetti A, Caponnetto S, Marchetti P, Nuti M. Exploratory Pilot Study of Circulating Biomarkers in Metastatic Renal Cell Carcinoma. Cancers (Basel) 2020; 12:cancers12092620. [PMID: 32937860 PMCID: PMC7563741 DOI: 10.3390/cancers12092620] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary The identification of biomarkers in response to therapeutic treatment is one of the main objectives of personalized oncology. Predictive biomarkers are particularly relevant for oncologists challenged by the busy scenario of possible therapeutic options in mRCC patients, including immunotherapy and TKIs. In fact the activation of the immune system can determine the outcome and success of the different therapeutic strategies. In this study we evaluated changes in the immune system of TKI mRCC-treated patients defining immunological profiles related to response characterized by specific biomarkers. The validation of the proposed immune portrait to an extended number of patients could allow characterization and selection of responsive and non-responsive patients from the beginning of the therapeutic process. Abstract With the introduction of immune checkpoint inhibitors (ICIs) and next-generation vascular endothelial growth factor receptor–tyrosine kinase inhibitors (VEGFR–TKIs), the survival of patients with advanced renal cell carcinoma (RCC) has improved remarkably. However, not all patients have benefited from treatments, and to date, there are still no validated biomarkers that can be included in the therapeutic algorithm. Thus, the identification of predictive biomarkers is necessary to increase the number of responsive patients and to understand the underlying immunity. The clinical outcome of RCC patients is, in fact, associated with immune response. In this exploratory pilot study, we assessed the immune effect of TKI therapy in order to evaluate the immune status of metastatic renal cell carcinoma (mRCC) patients so that we could define a combination of immunological biomarkers relevant to improving patient outcomes. We profiled the circulating levels in 20 mRCC patients of exhausted/activated/regulatory T cell subsets through flow cytometry and of 14 immune checkpoint-related proteins and 20 inflammation cytokines/chemokines using multiplex Luminex assay, both at baseline and during TKI therapy. We identified the CD3+CD8+CD137+ and CD3+CD137+PD1+ T cell populations, as well as seven soluble immune molecules (i.e., IFNγ, sPDL2, sHVEM, sPD1, sGITR, sPDL1, and sCTLA4) associated with the clinical responses of mRCC patients, either modulated by TKI therapy or not. These results suggest an immunological profile of mRCC patients, which will help to improve clinical decision-making for RCC patients in terms of the best combination of strategies, as well as the optimal timing and therapeutic sequence.
Collapse
Affiliation(s)
- Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (A.R.); (M.N.)
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (A.R.); (M.N.)
- Correspondence: ; Tel.: +39-064-997-3025
| | - Alessandra Di Filippo
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (A.R.); (M.N.)
| | - Andrea Botticelli
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Alain Gelibter
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Fabio Calabrò
- Division of Medical Oncology B, San Camillo Forlanini Hospital Rome, 00149 Rome, Italy;
| | - Ernesto Rossi
- Department of Medical Oncology, Fondazione Policlinico A.Gemelli Rome, 00168 Rome, Italy; (E.R.); (G.S.)
| | - Giovanni Schinzari
- Department of Medical Oncology, Fondazione Policlinico A.Gemelli Rome, 00168 Rome, Italy; (E.R.); (G.S.)
| | - Federica Urbano
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Giulia Pomati
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Simone Scagnoli
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (A.R.); (M.N.)
| | - Salvatore Caponnetto
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
| | - Paolo Marchetti
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (A.B.); (A.G.); (F.U.); (G.P.); (S.S.); (S.C.); (P.M.)
- Division of Oncology, Department of Clinical and Molecular Medicine, Ospedale Sant’Andrea, “Sapienza” University of Rome, 00189 Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, “Sapienza” University of Rome, 00161 Rome, Italy; (I.G.Z.); (A.D.F.); (A.R.); (M.N.)
| |
Collapse
|
11
|
Claus C, Ferrara C, Xu W, Sam J, Lang S, Uhlenbrock F, Albrecht R, Herter S, Schlenker R, Hüsser T, Diggelmann S, Challier J, Mössner E, Hosse RJ, Hofer T, Brünker P, Joseph C, Benz J, Ringler P, Stahlberg H, Lauer M, Perro M, Chen S, Küttel C, Bhavani Mohan PL, Nicolini V, Birk MC, Ongaro A, Prince C, Gianotti R, Dugan G, Whitlow CT, Solingapuram Sai KK, Caudell DL, Burgos-Rodriguez AG, Cline JM, Hettich M, Ceppi M, Giusti AM, Crameri F, Driessen W, Morcos PN, Freimoser-Grundschober A, Levitsky V, Amann M, Grau-Richards S, von Hirschheydt T, Tournaviti S, Mølhøj M, Fauti T, Heinzelmann-Schwarz V, Teichgräber V, Colombetti S, Bacac M, Zippelius A, Klein C, Umaña P. Tumor-targeted 4-1BB agonists for combination with T cell bispecific antibodies as off-the-shelf therapy. Sci Transl Med 2020; 11:11/496/eaav5989. [PMID: 31189721 DOI: 10.1126/scitranslmed.aav5989] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 05/16/2019] [Indexed: 01/08/2023]
Abstract
Endogenous costimulatory molecules on T cells such as 4-1BB (CD137) can be leveraged for cancer immunotherapy. Systemic administration of agonistic anti-4-1BB antibodies, although effective preclinically, has not advanced to phase 3 trials because they have been hampered by both dependency on Fcγ receptor-mediated hyperclustering and hepatotoxicity. To overcome these issues, we engineered proteins simultaneously targeting 4-1BB and a tumor stroma or tumor antigen: FAP-4-1BBL (RG7826) and CD19-4-1BBL. In the presence of a T cell receptor signal, they provide potent T cell costimulation strictly dependent on tumor antigen-mediated hyperclustering without systemic activation by FcγR binding. We could show targeting of FAP-4-1BBL to FAP-expressing tumor stroma and lymph nodes in a colorectal cancer-bearing rhesus monkey. Combination of FAP-4-1BBL with tumor antigen-targeted T cell bispecific (TCB) molecules in human tumor samples led to increased IFN-γ and granzyme B secretion. Further, combination of FAP- or CD19-4-1BBL with CEA-TCB (RG7802) or CD20-TCB (RG6026), respectively, resulted in tumor remission in mouse models, accompanied by intratumoral accumulation of activated effector CD8+ T cells. FAP- and CD19-4-1BBL thus represent an off-the-shelf combination immunotherapy without requiring genetic modification of effector cells for the treatment of solid and hematological malignancies.
Collapse
Affiliation(s)
- Christina Claus
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Claudia Ferrara
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Wei Xu
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Johannes Sam
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Sabine Lang
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Franziska Uhlenbrock
- University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Rosmarie Albrecht
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Sylvia Herter
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Ramona Schlenker
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Tamara Hüsser
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Sarah Diggelmann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - John Challier
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Ekkehard Mössner
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Ralf J Hosse
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Thomas Hofer
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Peter Brünker
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Catherine Joseph
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Jörg Benz
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Philippe Ringler
- University of Basel, Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Henning Stahlberg
- University of Basel, Center for Cellular Imaging and NanoAnalytics (C-CINA), Biozentrum, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Matthias Lauer
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Mario Perro
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Stanford Chen
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Christine Küttel
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Preethi L Bhavani Mohan
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Valeria Nicolini
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Martina Carola Birk
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Amandine Ongaro
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Christophe Prince
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Reto Gianotti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Gregory Dugan
- Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Christopher T Whitlow
- Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | - David L Caudell
- Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | - J Mark Cline
- Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Michael Hettich
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Maurizio Ceppi
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Anna Maria Giusti
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Flavio Crameri
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Wouter Driessen
- Roche Innovation Center Basel, pRED, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Peter N Morcos
- Roche Innovation Center New York, pRED, 430 E 29th St, New York, NY 10016, USA
| | - Anne Freimoser-Grundschober
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Victor Levitsky
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Maria Amann
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Sandra Grau-Richards
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | | | - Stella Tournaviti
- Roche Innovation Center Munich, pRED, Nonnenwald 2, 82377 Penzberg, Germany
| | - Michael Mølhøj
- Roche Innovation Center Munich, pRED, Nonnenwald 2, 82377 Penzberg, Germany
| | - Tanja Fauti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | | | - Volker Teichgräber
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Sara Colombetti
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Marina Bacac
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Alfred Zippelius
- University of Basel, Department of Biomedicine, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Christian Klein
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland
| | - Pablo Umaña
- Roche Innovation Center Zurich, Roche Pharmaceutical Research and Early Development (pRED), Wagistrasse 10, 8952 Schlieren, Switzerland.
| |
Collapse
|
12
|
Qu QX, Zhu XY, Du WW, Wang HB, Shen Y, Zhu YB, Chen C. 4-1BB Agonism Combined With PD-L1 Blockade Increases the Number of Tissue-Resident CD8+ T Cells and Facilitates Tumor Abrogation. Front Immunol 2020; 11:577. [PMID: 32391001 PMCID: PMC7193033 DOI: 10.3389/fimmu.2020.00577] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/12/2020] [Indexed: 12/23/2022] Open
Abstract
Although the milestone discovery of immune checkpoint blockade (ICB) has been translated into clinical practice, only a fraction of patients can benefit from it with durable responses and subsequent long-term survival. Here, we tested the anti-tumor effect of combining PD-L1 blockade with 4-1BB costimulation in 3LL and 4T1.2 murine tumor models. Dual treatment induced further tumor regression and enhanced survival in tumor-bearing mice more so than PD-L1 and 4-1BB mAb alone. It was demonstrated that dual anti-PD-L1/anti-4-1BB immunotherapy increased the number of intratumoral CD103+CD8+ T cells and altered their distribution. Phenotypically, CD103+CD8+ T cells expressed a higher level of 4-1BB and PD-1 than their CD103− counterparts. Administration of PD-L1 mAb and 4-1BB mAb further increased the cytolytic capacity of CD103+CD8+ T cells. In vivo, CD103−CD8+ T cells could differentiate into CD103+CD8+ progeny cells. In a human setting, more CD8+ T cells differentiated into CD103+CD8+ T cells in the peripheral tumor region of lung cancer tissues than in the central tumor region. Collectively, infiltrated CD103+CD8+ T cells served as a potential effector T cell population. Combining 4-1BB agonism with PD-L1 blockade could increase tumor-infiltrated CD103+CD8+T cells, thereby facilitating tumor regression.
Collapse
Affiliation(s)
- Qiu-Xia Qu
- Clinical Immunology Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Xin-Yun Zhu
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wen-Wen Du
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hong-Bin Wang
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Shen
- Clinical Immunology Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Yi-Bei Zhu
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
| | - Cheng Chen
- Department of Respiratory and Critical Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
13
|
Targeting Negative and Positive Immune Checkpoints with Monoclonal Antibodies in Therapy of Cancer. Cancers (Basel) 2019; 11:cancers11111756. [PMID: 31717326 PMCID: PMC6895894 DOI: 10.3390/cancers11111756] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023] Open
Abstract
The immune checkpoints are regulatory molecules that maintain immune homeostasis in physiological conditions. By sending T cells a series of co-stimulatory or co-inhibitory signals via receptors, immune checkpoints can both protect healthy tissues from adaptive immune response and activate lymphocytes to remove pathogens effectively. However, due to their mode of action, suppressive immune checkpoints may serve as unwanted protection for cancer cells. To restore the functioning of the immune system and make the patient’s immune cells able to recognize and destroy tumors, monoclonal antibodies are broadly used in cancer immunotherapy to block the suppressive or to stimulate the positive immune checkpoints. In this review, we aim to present the current state of application of monoclonal antibodies in clinics, used either as single agents or in a combined treatment. We discuss the limitations of these therapies and possible problem-solving with combined treatment approaches involving both non-biological and biological agents. We also highlight the most promising strategies based on the use of monoclonal or bispecific antibodies targeted on immune checkpoints other than currently implemented in clinics.
Collapse
|
14
|
Ballesteros-Briones MC, Martisova E, Casales E, Silva-Pilipich N, Buñuales M, Galindo J, Mancheño U, Gorraiz M, Lasarte JJ, Kochan G, Escors D, Sanchez-Paulete AR, Melero I, Prieto J, Hernandez-Alcoceba R, Hervas-Stubbs S, Smerdou C. Short-Term Local Expression of a PD-L1 Blocking Antibody from a Self-Replicating RNA Vector Induces Potent Antitumor Responses. Mol Ther 2019; 27:1892-1905. [PMID: 31563534 DOI: 10.1016/j.ymthe.2019.09.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint blockade has shown anti-cancer efficacy, but requires systemic administration of monoclonal antibodies (mAbs), often leading to adverse effects. To avoid toxicity, mAbs could be expressed locally in tumors. We developed adeno-associated virus (AAV) and Semliki Forest virus (SFV) vectors expressing anti-programmed death ligand 1 (aPDL1) mAb. When injected intratumorally in MC38 tumors, both viral vectors led to similar local mAb expression at 24 h, diminishing quickly in SFV-aPDL1-treated tumors. However, SFV-aPDL1 induced >40% complete regressions and was superior to AAV-aPDL1, as well as to aPDL1 mAb given systemically or locally. SFV-aPDL1 induced abscopal effects and was also efficacious against B16-ovalbumin (OVA). The higher SFV-aPDL1 antitumor activity could be related to local upregulation of interferon-stimulated genes because of SFV RNA replication. This was confirmed by combining local SFV-LacZ administration and systemic aPDL1 mAb, which provided higher antitumor effects than each separated agent. SFV-aPDL1 promoted tumor-specific CD8 T cells infiltration in both tumor models. In MC38, SFV-aPDL1 upregulated co-stimulatory markers (CD137/OX40) in tumor CD8 T cells, and its combination with anti-CD137 mAb showed more pronounced antitumor effects than each single agent. These results indicate that local transient expression of immunomodulatory mAbs using non-propagative RNA vectors inducing type I interferon (IFN-I) responses represents a potent and safe approach for cancer treatment.
Collapse
Affiliation(s)
- Maria Cristina Ballesteros-Briones
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Eva Martisova
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Erkuden Casales
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Noelia Silva-Pilipich
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Maria Buñuales
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Javier Galindo
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Uxua Mancheño
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Marta Gorraiz
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Juan J Lasarte
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Grazyna Kochan
- Department of Oncology, Navarrabiomed-Biomedical Research Centre, IdiSNA, 31008 Pamplona, Spain
| | - David Escors
- Department of Oncology, Navarrabiomed-Biomedical Research Centre, IdiSNA, 31008 Pamplona, Spain
| | - Alfonso R Sanchez-Paulete
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain; Department of Immunology and Immunotherapy, Clinica Universidad de Navarra, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Jesus Prieto
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Ruben Hernandez-Alcoceba
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Division of Immunology and Immunotherapy, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain.
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra and Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain.
| |
Collapse
|
15
|
Grimmig T, Gasser M, Moench R, Zhu LJ, Nawalaniec K, Callies S, Wagner M, Polat B, Mothi SS, Luo Y, Ribas CM, Malafaia O, Hsiao LL, Waaga-Gasser AM. Expression of Tumor-mediated CD137 ligand in human colon cancer indicates dual signaling effects. Oncoimmunology 2019; 8:e1651622. [PMID: 31741755 PMCID: PMC6844327 DOI: 10.1080/2162402x.2019.1651622] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 07/01/2019] [Accepted: 07/31/2019] [Indexed: 12/17/2022] Open
Abstract
CD137-targeting immune therapy, which activates anti-tumor T effector cell responses, seems to be an attractive concept in clinical oncology. Recent evidence has demonstrated that tumor cells besides T cells and antigen-presenting cells are able to express CD137 and CD137L. Here we aimed to identify CD137/CD137L expression in established colon cancer cell lines and primary tumors (UICC stages I-IV) from patients with documented long-term follow-up. CD137/CD137L expression was highly upregulated in early to late-stage tumors while the inverse was observed in patient-derived peripheral blood mononuclear cells. High CD137L expression within primary tumors was mediated by tumor cells and significantly correlated with the occurrence of distant metastases and shortened survival in advanced stages of disease (UICC stage IV). Interestingly, induced tumor cell signaling via CD137L on its surface in vitro resulted in dual effects: (i) reduced tumor cell proliferation suggesting inhibitory signaling in all investigated cancers and (ii) increased epithelial-to-mesenchymal transition signaling events. Taken together CD137/CD137L expression was stage-dependently upregulated with shortened survival in patients with highly CD137L-expressing tumors. Our clinical and experimental data suggest that colon cancer cells predominantly express CD137L and thereby have negative impact on overall survival through a process of reverse signaling. Beside agonistic CD137 antibody therapy to foster T effector cell responses, CD137L-mediated intervention strategies may become instrumental to circumvent relapsed tumor growth through induced epithelial-to-mesenchymal transition and consecutive metastases formation.
Collapse
Affiliation(s)
- Tanja Grimmig
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Martin Gasser
- Department of Surgery I, University of Wuerzburg, Wuerzburg, Germany
| | - Romana Moench
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Lang-Jing Zhu
- Nephrology Department, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, PR. China.,Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karol Nawalaniec
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simone Callies
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Martin Wagner
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany
| | - Buelent Polat
- Department of Radiation, Oncology University of Wuerzburg, Wuerzburg, Germany
| | - Suraj Sarvode Mothi
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yueming Luo
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Carmen M Ribas
- Evangelical Medical School, Faculty University of Parana, Curitiba, Brazil
| | - Osvaldo Malafaia
- Evangelical Medical School, Faculty University of Parana, Curitiba, Brazil
| | - Li-Li Hsiao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana Maria Waaga-Gasser
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, Wuerzburg, Germany.,Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Guillerey C, Nakamura K, Pichler AC, Barkauskas D, Krumeich S, Stannard K, Miles K, Harjunpää H, Yu Y, Casey M, Doban AI, Lazar M, Hartel G, Smith D, Vuckovic S, Teng MW, Bergsagel PL, Chesi M, Hill GR, Martinet L, Smyth MJ. Chemotherapy followed by anti-CD137 mAb immunotherapy improves disease control in a mouse myeloma model. JCI Insight 2019; 5:125932. [PMID: 31194697 DOI: 10.1172/jci.insight.125932] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy holds promise for multiple myeloma (MM) patients but little is known about how MM-induced immunosuppression influences response to therapy. Here, we investigated the impact of disease progression on immunotherapy efficacy in the Vk*MYC mouse model. Treatment with agonistic anti-CD137 (4-1BB) mAbs efficiently protected mice when administered early but failed to contain MM growth when delayed more than three weeks after Vk*MYC tumor cell challenge. The quality of CD8+ T cell response to CD137 stimulation was not altered by the presence of MM, but CD8+ T cell numbers were profoundly reduced at the time of treatment. Our data suggest that an insufficient ratio of CD8+ T cells over MM cells (CD8/MM) accounts for the loss of anti-CD137 mAb efficacy. We established serum M-protein levels prior to therapy as a predictive factor of response. Moreover, we developed an in silico model to capture the dynamic interactions between CD8+ T cells and MM cells. Finally, we explored two methods to improve the CD8/MM ratio: anti-CD137 mAb immunotherapy combined with Treg-depletion or administered after chemotherapy treatment with cyclophosphamide or melphalan efficiently reduced MM burden and prolonged survival. Altogether, our data indicate that consolidation treatment with anti-CD137 mAbs might prevent MM relapse.
Collapse
Affiliation(s)
- Camille Guillerey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia.,Cancer Immunotherapies Laboratory, Mater Research Institute, The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Kyohei Nakamura
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Andrea C Pichler
- Cancer Research Center of Toulouse, INSERM UMR 1037, Toulouse, France
| | - Deborah Barkauskas
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Sophie Krumeich
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kimberley Stannard
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kim Miles
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Heidi Harjunpää
- School of Medicine, The University of Queensland, Herston, Queensland, Australia.,Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Yuan Yu
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mika Casey
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | | | - Mircea Lazar
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | | - Slavica Vuckovic
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Multiple Myeloma Research Group, Institute of Haematology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Michele Wl Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - P Leif Bergsagel
- Comprehensive Cancer Center, Mayo Clinic, Scottsdale, Arizona, USA
| | - Marta Chesi
- Comprehensive Cancer Center, Mayo Clinic, Scottsdale, Arizona, USA
| | - Geoffrey R Hill
- Bone Marrow Transplantation Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ludovic Martinet
- Cancer Research Center of Toulouse, INSERM UMR 1037, Toulouse, France
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
17
|
Hinner MJ, Aiba RSB, Jaquin TJ, Berger S, Dürr MC, Schlosser C, Allersdorfer A, Wiedenmann A, Matschiner G, Schüler J, Moebius U, Rothe C, Matis L, Olwill SA. Tumor-Localized Costimulatory T-Cell Engagement by the 4-1BB/HER2 Bispecific Antibody-Anticalin Fusion PRS-343. Clin Cancer Res 2019; 25:5878-5889. [DOI: 10.1158/1078-0432.ccr-18-3654] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/14/2019] [Accepted: 05/22/2019] [Indexed: 11/16/2022]
|
18
|
Aravinth SP, Rajendran S, Li Y, Wu M, Yi Wong AH, Schwarz H. Epstein-Barr virus-encoded LMP1 induces ectopic CD137 expression on Hodgkin and Reed-Sternberg cells via the PI3K-AKT-mTOR pathway. Leuk Lymphoma 2019; 60:2697-2704. [PMID: 31058559 DOI: 10.1080/10428194.2019.1607330] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CD137 is a potent co-stimulatory molecule on activated T cells, and its ligand (CD137L) is expressed on antigen presenting cells (APC). Ectopic expression of CD137 has been identified on Hodgkin Reed-Sternberg (HRS) cells, the malignant cells in Hodgkin Lymphoma (HL), and CD137 on HRS cells was found to support growth of HRS cells and escape from immune surveillance. HRS cells are mostly derived from B cells, which poses the question of how B cells acquire ectopic CD137 expression during the transformation process. HL is associated with Epstein-Barr virus (EBV) infection. We show that the EBV latent membrane protein 1 (LMP1) induces expression of CD137 in HRS cell lines. In a HL tissue microarray, 96% of the CD137-positive HL cases stained positive for LMP1. LMP1 utilizes the PI3K-AKT-mTOR pathway for inducing CD137 expression. These findings support the role of EBV in HL pathogenesis.
Collapse
Affiliation(s)
- Sneha Priya Aravinth
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sakthi Rajendran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Yating Li
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Meihui Wu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Anna Hiu Yi Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Wang D, Lin J, Yang X, Long J, Bai Y, Yang X, Mao Y, Sang X, Seery S, Zhao H. Combination regimens with PD-1/PD-L1 immune checkpoint inhibitors for gastrointestinal malignancies. J Hematol Oncol 2019; 12:42. [PMID: 31014381 PMCID: PMC6480748 DOI: 10.1186/s13045-019-0730-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/02/2019] [Indexed: 12/24/2022] Open
Abstract
Gastrointestinal (GI) malignant neoplasms have a high global incidence and treatment prospects for patients with advanced GI tumors are dismal. PD-1/PD-L1 inhibitors emerged as a frontline treatment for several types of cancer. However, the shortcomings of PD-1/PD-L1 inhibitors have been observed, including low objective response rates and acquired tumor resistance, especially in patients receiving PD-1/PD-L1 inhibitors as a single treatment. Accumulating evidence from clinical trials increasingly suggests that combined immunotherapies enhance therapeutic responses in patients with malignances, especially for GI tumors which have a complex matrix, and significant molecular and immunological differences. Preclinical and clinical studies suggest there are advantages to combined immunological regimens, which represents the next logical step in this field, although further research is necessary. This literature review explores the current limitations of monotherapies, before critically discussing the rationale behind combination regimens. Then, we provide a summary of the clinical applications for gastrointestinal cancers.
Collapse
Affiliation(s)
- Dongxu Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaobo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Samuel Seery
- Department of Humanities, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
20
|
Nong J, Wang J, Gao X, Zhang Q, Yang B, Yan Z, Wang X, Yi L, Wang Q, Gao Y, Hu A, Qin N, Wei P, Zhang H, Zhang S. Circulating CD137 + CD8 + T cells accumulate along with increased functional regulatory T cells and thoracic tumour burden in lung cancer patients. Scand J Immunol 2019; 89:e12765. [PMID: 30921475 DOI: 10.1111/sji.12765] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 03/13/2019] [Accepted: 03/18/2019] [Indexed: 01/04/2023]
Abstract
CD137 is a promising target for immunostimulation strategies against cancer. Previous studies showed that CD137+ CD8+ T cells are enriched in antitumour effector T cells in both preclinical tumour models and cancer patients, but to date, such T cells in the blood of lung cancer patients have not been sufficiently investigated. In this study, circulating antigen-activated CD8+ T cell subsets, identified as CD137+ CD8+ or PD-1+ (programmed cell death protein 1) CD8+ , and regulatory T cells (Treg), identified as CD4+ CD25+ CD127low/- , in 40 untreated lung cancer patients and in 49 age- and sex-matched healthy controls (HCs) were assessed by flow cytometry. Results were evaluated for associations with lung cancer patient clinical characteristics. Correlations between antigen-activated CD8+ T cells and effector Treg (CTLA-4+ [cytotoxic T-lymphocyte antigen 4] CD4+ CD25+ CD127low/- ) were also investigated. Higher percentages of PD-1+ , CD137+ and PD-1+ CD137+ amongst CD8+ T cells were observed in lung cancer patients compared with HCs. The percentages of CD137+ CD8+ and PD-1+ CD137+ CD8+ T cell subsets amongst CD8+ T cells were positively correlated with thoracic tumour burden and were strongly positively correlated with the percentage of effector Treg subset. Smoking patients harboured higher percentages of the PD-1+ CD8+ T cell subset compared with non-smoking patients. This study demonstrated that circulating antigen-activated CD8+ T cells accumulated in lung cancer patients along with increased effector Treg and thoracic tumour burden. These findings aid a better understanding of immune-host interactions in lung cancer patients using peripheral blood, and further support immunotherapeutic intervention strategies using combination therapy for differential control of Treg and activation of tumour-specific effector T cells.
Collapse
Affiliation(s)
- Jingying Nong
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Xin Gao
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Qi Zhang
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Bin Yang
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Zhuohong Yan
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Ling Yi
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Qunhui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Yuan Gao
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Aimin Hu
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Na Qin
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Panjian Wei
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Key Laboratory for Drug Resistance Tuberculosis Research, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Shucai Zhang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| |
Collapse
|
21
|
Strome AL, Zhang X, Strome SE. The evolving role of immuno-oncology for the treatment of head and neck cancer. Laryngoscope Investig Otolaryngol 2019; 4:62-69. [PMID: 30828620 PMCID: PMC6383301 DOI: 10.1002/lio2.235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/08/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Monoclonal antibodies (mAbs) that target immune co‐signaling pathways have the potential to enable immune mediated tumor eradication. While early adoption of these agents for the treatment of advanced squamous cell carcinoma of the head and neck (SCCHN) has produced some astounding clinical successes, the majority of patients fail to respond to therapy. The purpose of this review is to first provide a broad overview of the immuno‐oncology (I‐O) landscape and to then focus on the current status of mAb‐based I‐O (mAb:I‐O) for the treatment of SCCHN, with particular attention to the development of strategies for improving treatment responses.
Collapse
Affiliation(s)
| | - Xiaoyu Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery University of Maryland School of Medicine Baltimore Maryland
| | - Scott E Strome
- Department of Otorhinolaryngology-Head and Neck Surgery University of Maryland School of Medicine Baltimore Maryland.,College of Medicine University of Tennessee Health Science Center Memphis Tennessee
| |
Collapse
|
22
|
Structure of the 4-1BB/4-1BBL complex and distinct binding and functional properties of utomilumab and urelumab. Nat Commun 2018; 9:4679. [PMID: 30410017 PMCID: PMC6224509 DOI: 10.1038/s41467-018-07136-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 10/09/2018] [Indexed: 12/20/2022] Open
Abstract
4-1BB (CD137, TNFRSF9) is an inducible costimulatory receptor expressed on activated T cells. Clinical trials of two agonist antibodies, utomilumab (PF-05082566) and urelumab (BMS-663513), are ongoing in multiple cancer indications, and both antibodies demonstrate distinct activities in the clinic. To understand these differences, we solved structures of the human 4-1BB/4-1BBL complex, the 4-1BBL trimer alone, and 4-1BB bound to utomilumab or urelumab. The 4-1BB/4-1BBL complex displays a unique interaction between receptor and ligand when compared with other TNF family members. Furthermore, our ligand-only structure differs from previously published data. Utomilumab, a ligand-blocking antibody, binds 4-1BB between CRDs 3 and 4. In contrast, urelumab binds 4-1BB CRD-1, away from the ligand binding site. Finally, cell-based assays demonstrate utomilumab is a milder agonist than urelumab. Collectively, our data provide a deeper understanding of the 4-1BB signaling complex, providing a template for future development of next generation 4-1BB targeted biologics.
Collapse
|
23
|
Attili I, Karachaliou N, Bonanno L, Berenguer J, Bracht J, Codony-Servat J, Codony-Servat C, Ito M, Rosell R. STAT3 as a potential immunotherapy biomarker in oncogene-addicted non-small cell lung cancer. Ther Adv Med Oncol 2018; 10:1758835918763744. [PMID: 29636826 PMCID: PMC5888808 DOI: 10.1177/1758835918763744] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/15/2018] [Indexed: 12/27/2022] Open
Abstract
Immune checkpoint blockade has modified the treatment landscape for many types of tumors, including lung cancer. Still our knowledge on the biology of the interaction between tumor cells and the microenvironment is limited, preventing the optimal use of these new compounds and the maximum benefit that the patients can derive from them. We have actively worked on the role of STAT3, a transcriptional factor that causes innate resistance to targeted therapies in oncogene-addicted tumors. In this short review we take the opportunity to express our opinion and review existing knowledge on the immune role of STAT3 and the possible implications that this may have for the discovery of new biomarkers to predict response to immunotherapy, as well as new partners to combine with and increase the efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ilaria Attili
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 53, Padova 35128, Italy
| | - Niki Karachaliou
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Instituto Oncológico Dr Rosell (IOR), University Hospital Sagrat Cor, Barcelona, Spain
| | | | - Jordi Berenguer
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Jillian Bracht
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Jordi Codony-Servat
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Carles Codony-Servat
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Masaoki Ito
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Rafael Rosell
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Instituto Oncológico Dr Rosell (IOR), Quirón-Dexeus University Institute, Barcelona, Spain
- Institut d’Investigació en Ciències Germans Trias i Pujol, Badalona, Spain
- Institut Català d’Oncologia, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
24
|
Karachaliou N, Rosell R. Science and biology drives the immune system to cure lung cancer patients: a revolution but not without challenges. Ther Adv Med Oncol 2018; 10:1758835918763723. [PMID: 29619092 PMCID: PMC5871042 DOI: 10.1177/1758835918763723] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/15/2018] [Indexed: 12/26/2022] Open
Affiliation(s)
- Niki Karachaliou
- Instituto Oncológico Dr Rosell (IOR), University Hospital Sagrat Cor, QuirónSalud Group, Viladomat 288, 08029 Barcelona, Spain
| | - Rafael Rosell
- Instituto Oncológico Dr Rosell (IOR), Quirón-Dexeus University Institute, Barcelona, Spain; Institut d'Investigació en Ciències Germans Trias i Pujol, Badalona, Spain; Institut Català d'Oncologia, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
25
|
Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol 2018; 11:39. [PMID: 29544515 PMCID: PMC5856308 DOI: 10.1186/s13045-018-0582-8] [Citation(s) in RCA: 541] [Impact Index Per Article: 90.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/01/2018] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoints consist of inhibitory and stimulatory pathways that maintain self-tolerance and assist with immune response. In cancer, immune checkpoint pathways are often activated to inhibit the nascent anti-tumor immune response. Immune checkpoint therapies act by blocking or stimulating these pathways and enhance the body's immunological activity against tumors. Cytotoxic T lymphocyte-associated molecule-4 (CTLA-4), programmed cell death receptor-1 (PD-1), and programmed cell death ligand-1(PD-L1) are the most widely studied and recognized inhibitory checkpoint pathways. Drugs blocking these pathways are currently utilized for a wide variety of malignancies and have demonstrated durable clinical activities in a subset of cancer patients. This approach is rapidly extending beyond CTLA-4 and PD-1/PD-L1. New inhibitory pathways are under investigation, and drugs blocking LAG-3, TIM-3, TIGIT, VISTA, or B7/H3 are being investigated. Furthermore, agonists of stimulatory checkpoint pathways such as OX40, ICOS, GITR, 4-1BB, CD40, or molecules targeting tumor microenvironment components like IDO or TLR are under investigation. In this article, we have provided a comprehensive review of immune checkpoint pathways involved in cancer immunotherapy, and discuss their mechanisms and the therapeutic interventions currently under investigation in phase I/II clinical trials. We also reviewed the limitations, toxicities, and challenges and outline the possible future research directions.
Collapse
Affiliation(s)
| | - Bhagirathbhai Dholaria
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
- Present Address: Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Aixa E Soyano
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Saranya Chumsri
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
26
|
Van Blarcom T, Lindquist K, Melton Z, Cheung WL, Wagstrom C, McDonough D, Valle Oseguera C, Ding S, Rossi A, Potluri S, Sundar P, Pitts S, Sirota M, Galindo Casas M, Yan Y, Jones J, Roe-Zurz Z, Srivatsa Srinivasan S, Zhai W, Pons J, Rajpal A, Chaparro-Riggers J. Productive common light chain libraries yield diverse panels of high affinity bispecific antibodies. MAbs 2017; 10:256-268. [PMID: 29227213 PMCID: PMC5825193 DOI: 10.1080/19420862.2017.1406570] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The commercial success of bispecific antibodies generally has been hindered by the complexities associated with generating appropriate molecules for both research scale and large scale manufacturing purposes. Bispecific IgG (BsIgG) based on two antibodies that use an identical common light chain can be combined with a minimal set of Fc mutations to drive heavy chain heterodimerization in order to address these challenges. However, the facile generation of common light chain antibodies with properties similar to traditional monoclonal antibodies has not been demonstrated and they have only been used sparingly. Here, we describe the design of a synthetic human antibody library based on common light chains to generate antibodies with biochemical and biophysical properties that are indistinguishable to traditional therapeutic monoclonal antibodies. We used this library to generate diverse panels of well-behaved, high affinity antibodies toward a variety of epitopes across multiple antigens, including mouse 4-1BB, a therapeutically important T cell costimulatory receptor. Over 200 BsIgG toward 4-1BB were generated using an automated purification method we developed that enables milligram-scale production of BsIgG. This approach allowed us to identify antibodies with a wide range of agonistic activity that are being used to further investigate the therapeutic potential of antibodies targeting one or more epitopes of 4-1BB.
Collapse
Affiliation(s)
- Thomas Van Blarcom
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Kevin Lindquist
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Zea Melton
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Wai Ling Cheung
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Chris Wagstrom
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Dan McDonough
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Cendy Valle Oseguera
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Sheng Ding
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Andrea Rossi
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Shobha Potluri
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Purnima Sundar
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Steven Pitts
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Marina Sirota
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Meri Galindo Casas
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Yu Yan
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Jeffrey Jones
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Zygy Roe-Zurz
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | | | - Wenwu Zhai
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Jaume Pons
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | - Arvind Rajpal
- a Oncology Research and Development , Pfizer Inc. , South San Francisco , CA , USA
| | | |
Collapse
|
27
|
Liu B, Song Y, Liu D. Clinical trials of CAR-T cells in China. J Hematol Oncol 2017; 10:166. [PMID: 29058636 PMCID: PMC5651613 DOI: 10.1186/s13045-017-0535-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Novel immunotherapeutic agents targeting tumor-site microenvironment are revolutionizing cancer therapy. Chimeric antigen receptor (CAR)-engineered T cells are widely studied for cancer immunotherapy. CD19-specific CAR-T cells, tisagenlecleucel, have been recently approved for clinical application. Ongoing clinical trials are testing CAR designs directed at novel targets involved in hematological and solid malignancies. In addition to trials of single-target CAR-T cells, simultaneous and sequential CAR-T cells are being studied for clinical applications. Multi-target CAR-engineered T cells are also entering clinical trials. T cell receptor-engineered CAR-T and universal CAR-T cells represent new frontiers in CAR-T cell development. In this study, we analyzed the characteristics of CAR constructs and registered clinical trials of CAR-T cells in China and provided a quick glimpse of the landscape of CAR-T studies in China.
Collapse
Affiliation(s)
- Bingshan Liu
- School of Basic Medical Sciences and The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China.,Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China
| | - Yongping Song
- Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China.
| | - Delong Liu
- Henan Cancer Hospital and The Affiliated Cancer Hospital of Zhengzhou University, 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|