1
|
Salehi S, Ghaderi H, Habibi-Anbouhi M, Shoari A, Hassanzadeh Eskafi A, Sabouri A, Hosseininejad-Chafi M, Ashja Ardalan A, Ramezani B, Kazemi-Lomedasht F, Behdani M. Tumor Suppression by PD-1/PD-L1 Interaction Blockage in Mice Model. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 21:e132329. [PMID: 36896323 PMCID: PMC9990516 DOI: 10.5812/ijpr-132329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/26/2022] [Accepted: 01/02/2023] [Indexed: 03/07/2023]
Abstract
Background Overexpression of programmed cell death ligand 1 (PD-L1) in tumor cells and subsequent interaction with the programmed cell death protein 1 (PD-1) in tumor-infiltrating T cells cause an immune evasion of the tumor from cytotoxic T-cells. Therefore, inhibiting such interaction by a recombinant PD-1 can hinder tumor growth and extend the survival rate. Methods The mouse extracellular domain of PD-1 (mPD-1) was expressed in E. coli BL21 (DE3) strain and purified using nickel affinity chromatography. The binding ability of the purified protein to human PD-L1 was studied using ELISA. Finally, the tumor-bearing mice were used to evaluate the potential antitumor effect. Results The recombinant mPD-1 showed a significant binding capacity to human PD-L1 at the molecular level. The tumor size significantly decreased in the tumor-bearing mice after the intra-tumoral injections of mPD-1. Moreover, the survival rate increased significantly after eight weeks of monitoring. The histopathology revealed the necrosis in the tumor tissue of the control group compared to the mPD-1 received mice. Conclusions Our outcomes propose that interaction blockade between PD-1 and PD-L1 is a promising approach for targeted tumor therapy.
Collapse
Affiliation(s)
- Shima Salehi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Hajarossadat Ghaderi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | | | - Alireza Shoari
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
- Corresponding Author: Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran.
| | - Ayda Hassanzadeh Eskafi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Sabouri
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Hosseininejad-Chafi
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Arghavan Ashja Ardalan
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Behzad Ramezani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
- Zoonoses Research Center, Pasteur Institute of Iran, Amol, Iran
- Corresponding Author: Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
2
|
Geng S, Xu R, Huang X, Li M, Deng C, Lai P, Wang Y, Wu P, Chen X, Weng J, Du X. Dynamics of PD-1 expression are associated with treatment efficacy and prognosis in patients with intermediate/high-risk myelodysplastic syndromes under hypomethylating treatment. Front Immunol 2022; 13:950134. [PMID: 36003379 PMCID: PMC9393298 DOI: 10.3389/fimmu.2022.950134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Hypomethylating agents (HMAs) are widely used in patients with higher-risk MDS not eligible for stem cell transplantation. However, the general response rate by HMAs is lesser than 50% in MDS patients, while the relapse rate is high. Emerging evidence indicates that demethylating effects committed by HMAs may facilitate the up-regulation of a range of immune checkpoints or cancer suppressor genes in patients with MDS, among which the programmed death protein 1 (PD-1) and its ligands are demonstrated to be prominent and may contribute to treatment failure and early relapse. Although results from preliminary studies with a limited number of enrolled patients indicate that combined administration of PD-1 inhibitor may yield extra therapeutic benefit in some MDS patients, identifications of this subgroup of patients and optimal timing for the anti-PD-1 intervention remain significant challenges. Dynamics of immune checkpoints and associated predictive values during HMA-treatment cycles remained poorly investigated. In this present study, expression levels of immune checkpoints PD-1 and its ligands PD-L1 and PD-L2 were retrospectively analyzed by quantitative PCR (Q-PCR) in a total of 135 myelodysplastic syndromes (MDS) cohort with higher-risk stratification. The prognostic value of dynamics of these immune checkpoints during HMA cycles was validated in two independent prospective cohorts in our center (NCT01599325 and NCT01751867). Our data revealed that PD-1 expression was significantly higher than that in younger MDS patients (age ≤ 60) and MDS with lower IPSS risk stratification (intermediate risk-1). A significantly up-regulated expression of PD-1 was seen during the first four HMA treatment cycles in MDS patients, while similar observation was not seen concerning the expression of PD-L1 or PD-L2. By utilizing binary logistic regression and receiver operating characteristic (ROC) models, we further identified that higher or equal to 75.9 PD-1 expressions after 2 cycles of HMA treatment is an independent negative prognostic factor in predicting acute myeloid leukemia (AML) transformation and survival. Collectively, our data provide rationales for monitoring the expression of PD-1 during HMA treatment cycles, a higher than 75.9 PD-1 expression may identify patients who will potentially benefit from the combined therapy of HMA and PD-1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xin Du
- *Correspondence: Xin Du, ; ; Jianyu Weng,
| |
Collapse
|
3
|
Moe A, Liow E, Redfern A, Swarbrick N, Ferguson T, Davis ID, Hayne D. A phase I open label dose-escalation study to evaluate the tolerability, safety and immunological efficacy of sub-urothelial durvalumab injection in adults with muscle-invasive or high-risk non-muscle-invasive bladder cancer (SUBDUE-1, SUB-urothelial DUrvalumab injection-1 study): clinical trial protocol. BJU Int 2021; 128 Suppl 1:9-17. [PMID: 33960102 DOI: 10.1111/bju.15365] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
OBJECTIVES This article presents the clinical trial protocol for a phase I open label dose-escalation study to evaluate the tolerability, safety and immunological efficacy of sub-urothelial durvalumab injection in adults with muscle-invasive or high-risk non-muscle-invasive bladder cancer (NMIBC), the SUB-urothelial DUrvalumab injection-1 study (SUBDUE-1). The primary objectives of this study are to assess the safety of sub-urothelial injection of durvalumab using patient reported outcome measures and observed local or systemic adverse events. The secondary objectives are to examine the local immunological efficacy of sub-urothelial administration of durvalumab. PATIENTS AND METHODS The SUBDUE-1 trial will include adult patients with either high-risk NMIBC or MIBC, who are scheduled for radical cystectomy or who have refused or are unsuitable for systemic neoadjuvant chemotherapy. Three fixed total dose levels of durvalumab (25, 75, 150 mg) will be studied to identify a dose suitable to be taken forward into phase II trials. The primary endpoint is to evaluate the safety and tolerability of the trial intervention in terms of the incidence and severity of adverse events and the potential establishment of dose-limiting toxicities. The secondary efficacy endpoints include rates of pT0 status at resection, lymph node status, as well as the change in distribution of tumour-infiltrating lymphocytes and tumour-activated macrophages between pre- and post-injection bladder biopsies. Translational studies will focus on bladder tumour molecular sub-typing, immune infiltrate characterisation, and immune checkpoint protein expression relative to efficacy end-points. OUTCOME AND SIGNIFICANCE If proven safe and effective, this novel strategy comprising sub-urothelial durvalumab injections aimed at promoting an anti-tumour immune reaction, will provide additional treatment options for reducing tumour recurrence and progression in treatment-naïve patients with high-risk NMIBC or in patients with bacille Calmette-Guérin-refractory NMIBC. Local administration of durvalumab may be associated with a reduced rate of immunological side-effects and lower costs when compared to systemic delivery.
Collapse
Affiliation(s)
- Andrew Moe
- University of Western Australia (UWA) Medical School, University of Western Australia, Perth, WA, Australia.,Urology and Medical Oncology Departments, South Metropolitan Health Service, Perth, WA, Australia.,Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, NSW, Australia
| | - Elizabeth Liow
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, NSW, Australia
| | - Andrew Redfern
- University of Western Australia (UWA) Medical School, University of Western Australia, Perth, WA, Australia.,Urology and Medical Oncology Departments, South Metropolitan Health Service, Perth, WA, Australia.,Cancer Division, Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Nicole Swarbrick
- Department of Anatomical Pathology, PathWest Laboratory Medicine WA, Perth, WA, Australia.,Division of Pathology and Laboratory Medicine, UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Tom Ferguson
- Urology and Medical Oncology Departments, South Metropolitan Health Service, Perth, WA, Australia
| | - Ian D Davis
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, NSW, Australia.,Eastern Health Clinical School, Monash University, Melbourne, Vic., Australia.,Department of Oncology, Eastern Health, Melbourne, Vic., Australia
| | - Dickon Hayne
- University of Western Australia (UWA) Medical School, University of Western Australia, Perth, WA, Australia.,Urology and Medical Oncology Departments, South Metropolitan Health Service, Perth, WA, Australia.,Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, NSW, Australia
| |
Collapse
|
4
|
Effect of the hairpin structure of peptide inhibitors on the blockade of PD-1/PD-L1 axis. Biochem Biophys Res Commun 2020; 527:453-457. [PMID: 32336542 DOI: 10.1016/j.bbrc.2020.04.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 11/20/2022]
Abstract
Blockade of the PD-1/PD-L1 axis using antibody drugs has been a clinically efficacious immunotherapy in cancer treatment. However, studies on peptide inhibitors blocking the interaction between PD-1/PD-L1 in cancer treatment in clinical practice have not yet been reported. In this study, a series of peptide inhibitors were synthesized based on a continuous sequence of 14 amino acids from PD-L1 and suitable modifications to form a hairpin structure. The effect of inhibitors on the blockage of PD-1/PD-L1 by increasing the stability of the hairpin structure was determined using BLI and co-culture models. The results showed that increasing the stability of the hairpin improved the affinity of inhibitors to PD-1 and increased IL-2 secretion. Therefore, modifying the hairpin structure of peptide inhibitors may be a useful approach to block the interaction between PD-1 and PD-L1.
Collapse
|
5
|
Rossa C, D'Silva NJ. Non-murine models to investigate tumor-immune interactions in head and neck cancer. Oncogene 2019; 38:4902-4914. [PMID: 30872793 PMCID: PMC6586515 DOI: 10.1038/s41388-019-0776-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 12/18/2022]
Abstract
The immune response has important roles in the biology of solid tumors, including oncogenesis, tumor growth, invasion and metastasis, and response to treatment. Improved understanding of tumor-immune system interactions has provided promising therapeutic options that are based on the rescue and enhancement of the anti-tumoral host response. Immune-based treatments have been approved for clinical use in various types of cancer, including head and neck cancer (HNC); other strategies involving combination therapies are currently in development. These novel therapies were developed based on knowledge derived from in vitro, in silico, and in vivo pre-clinical studies. However, clinical trials seldom replicate the efficacy observed in pre-clinical animal studies. This lack of correlation between pre-clinical studies and clinical trials may be related to limitations of the models used; which highlights the relevance of considering immune-related aspects of different pre-clinical models. Murine models are the most frequently used pre-clinical models of HNC and are discussed elsewhere. Non-murine models have characteristics that offer unique opportunities for the study of HNC etiology, therapeutic strategies, and tumor-immune system interactions. The current review focuses on immune-related aspects of non-murine models, including dog, cat, pig, zebrafish, and frog, that could be used to investigate tumor-immune interactions in HNC.
Collapse
Affiliation(s)
- Carlos Rossa
- Department of Diagnosis and Surgery, UNESP-State University of Sao Paulo, School of Dentistry at Araraquara, Araraquara, SP, Brazil. .,Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Nisha J D'Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA. .,Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Liang Z, Li Y, Tian Y, Zhang H, Cai W, Chen A, Chen L, Bao Y, Xiang B, Kan H, Li Y. High-affinity human programmed death-1 ligand-1 variant promotes redirected T cells to kill tumor cells. Cancer Lett 2019; 447:164-173. [PMID: 30677447 DOI: 10.1016/j.canlet.2019.01.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/17/2018] [Accepted: 01/14/2019] [Indexed: 12/31/2022]
Abstract
Tumor cells can escape immune surveillance through the programmed cell death protein 1 (PD-1) axis suppressing T cells. However, we recently demonstrated that high-affinity variants of soluble human programmed death-ligand 1 (shPD-L1) could diminish the suppression. We propose that in comparison to the wild-type shPD-L1, the further affinity enhancement will confer the molecule with opposite characteristics that augment T-cell activation and immunotherapeutic drug potential. In this study, a new shPD-L1 variant, L3C7c, has been generated to demonstrate ∼167 fold greater affinity than wild-type hPD-L1. The L3C7c-Fc fusion protein demonstrated completely opposite effects of conventional PD-1 axis by promoting redirected T-cell proliferation, activation and cytotoxicity in vitro, as being slightly better than that of anti-PD1-Ab (Pembrolizumab). Moreover, L3C7c-Fc was more effective than Pembrolizumab in enhancing redirected T cells' ability to suppress Mel624 melanoma growth in vivo. As a downsized L3C7c-Fc variant, L3C7v-Fc improved the anti-tumor efficacy in vivo when combined with dendritic cell vaccines. In conclusion, our studies demonstrate that high-affinity hPD-L1 variants could be developed as the next generation reagents for tumor immunotherapy based on the blockade of the PD-1 axis.
Collapse
Affiliation(s)
- Zhaoduan Liang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Yanyan Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China; School of Life Sciences, University of Science and Technology of China, No. 96, Jinzhai road, Hefei, Anhui province, China.
| | - Ye Tian
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Huanling Zhang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China; School of Life Sciences, University of Science and Technology of China, No. 96, Jinzhai road, Hefei, Anhui province, China.
| | - Wenxuan Cai
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Anan Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Lin Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Yifeng Bao
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| | - Bo Xiang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong province, China.
| | - Heping Kan
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong province, China.
| | - Yi Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kai Yuan Avenue, Science Park, Guangzhou, Guangdong province, China.
| |
Collapse
|
7
|
Schmohl JU, Nuebling T, Wild J, Kroell T, Kanz L, Salih HR, Schmetzer H. Expression of RANK-L and in part of PD-1 on blasts in patients with acute myeloid leukemia correlates with prognosis. Eur J Haematol 2016; 97:517-527. [DOI: 10.1111/ejh.12762] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Joerg Uwe Schmohl
- Section of Molecular Cancer Therapeutics; Therapeutic Radiology-Radiation Oncology; University of Minnesota; Masonic Cancer Center; Minneapolis MN USA
- Department for Hematology and Oncology; Med Dept II; University Hospital of Tuebingen; Tuebingen Germany
| | - Tina Nuebling
- Department for Hematology and Oncology; Med Dept II; University Hospital of Tuebingen; Tuebingen Germany
- Clinical Collaboration Unit Translational Immunology; German Cancer Consortium and German Cancer Research Center; Partner site Tuebingen; Department for Internal Medicine II; Eberhard Karls University Tuebingen; Tuebingen Germany
| | - Julia Wild
- Department for Hematology and Oncology; Med Dept II; University Hospital of Tuebingen; Tuebingen Germany
- Clinical Collaboration Unit Translational Immunology; German Cancer Consortium and German Cancer Research Center; Partner site Tuebingen; Department for Internal Medicine II; Eberhard Karls University Tuebingen; Tuebingen Germany
| | - Tanja Kroell
- Department for Hematopoetic Cell Transplantation; Med. Dept.III; University Hospital of Munich; Munich Germany
| | - Lothar Kanz
- Department for Hematology and Oncology; Med Dept II; University Hospital of Tuebingen; Tuebingen Germany
| | - Helmut Rainer Salih
- Department for Hematology and Oncology; Med Dept II; University Hospital of Tuebingen; Tuebingen Germany
- Clinical Collaboration Unit Translational Immunology; German Cancer Consortium and German Cancer Research Center; Partner site Tuebingen; Department for Internal Medicine II; Eberhard Karls University Tuebingen; Tuebingen Germany
| | - Helga Schmetzer
- Department for Hematopoetic Cell Transplantation; Med. Dept.III; University Hospital of Munich; Munich Germany
| |
Collapse
|
8
|
Stewart R, Morrow M, Hammond SA, Mulgrew K, Marcus D, Poon E, Watkins A, Mullins S, Chodorge M, Andrews J, Bannister D, Dick E, Crawford N, Parmentier J, Alimzhanov M, Babcook JS, Foltz IN, Buchanan A, Bedian V, Wilkinson RW, McCourt M. Identification and Characterization of MEDI4736, an Antagonistic Anti-PD-L1 Monoclonal Antibody. Cancer Immunol Res 2015; 3:1052-62. [PMID: 25943534 DOI: 10.1158/2326-6066.cir-14-0191] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/07/2015] [Indexed: 11/16/2022]
Abstract
Programmed cell-death 1 ligand 1 (PD-L1) is a member of the B7/CD28 family of proteins that control T-cell activation. Many tumors can upregulate expression of PD-L1, inhibiting antitumor T-cell responses and avoiding immune surveillance and elimination. We have identified and characterized MEDI4736, a human IgG1 monoclonal antibody that binds with high affinity and specificity to PD-L1 and is uniquely engineered to prevent antibody-dependent cell-mediated cytotoxicity. In vitro assays demonstrate that MEDI4736 is a potent antagonist of PD-L1 function, blocking interaction with PD-1 and CD80 to overcome inhibition of primary human T-cell activation. In vivo MEDI4736 significantly inhibits the growth of human tumors in a novel xenograft model containing coimplanted human T cells. This activity is entirely dependent on the presence of transplanted T cells, supporting the immunological mechanism of action for MEDI4736. To further determine the utility of PD-L1 blockade, an anti-mouse PD-L1 antibody was investigated in immunocompetent mice. Here, anti-mouse PD-L1 significantly improved survival of mice implanted with CT26 colorectal cancer cells. The antitumor activity of anti-PD-L1 was enhanced by combination with oxaliplatin, which resulted in increased release of HMGB1 within CT26 tumors. Taken together, our results demonstrate that inhibition of PD-L1 function can have potent antitumor activity when used as monotherapy or in combination in preclinical models, and suggest it may be a promising therapeutic approach for the treatment of cancer. MEDI4736 is currently in several clinical trials both alone and in combination with other agents, including anti-CTLA-4, anti-PD-1, and inhibitors of IDO, MEK, BRAF, and EGFR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Emily Dick
- MedImmune Ltd, Cambridge, United Kingdom
| | | | - Julie Parmentier
- Abbvie Inc, Worcester, Massachusetts. Previously AstraZeneca Ltd
| | - Marat Alimzhanov
- Acceleron Pharma, Inc, Cambridge, Massachusetts. Previously Astrazeneca Ltd
| | - John S Babcook
- CDRD, University of British Columbia, Vancouver, British Columbia, Canada. Previously Amgen Inc
| | - Ian N Foltz
- Amgen Inc, Burnaby, British Columbia, Canada
| | | | | | | | - Matthew McCourt
- Kymab Ltd, The Bennet Building, Babraham Research Campus, Cambridge, United Kingdom. Previously MedImmune Ltd
| |
Collapse
|
9
|
Mu N, Liu N, Hao Q, Xu Y, Li J, Li W, Wu S, Zhang C, Su H. Inhibition of mouse SP2/0 myeloma cell growth by the B7-H4 protein vaccine. BMB Rep 2015; 47:399-404. [PMID: 24314141 PMCID: PMC4163849 DOI: 10.5483/bmbrep.2014.47.7.168] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Indexed: 11/20/2022] Open
Abstract
B7-H4 is a member of B7 family of co-inhibitory molecules and B7-H4 protein is found to be overexpressed in many human cancers and which is usually associated with poor survival. In this study, we developed a therapeutic vaccine made from a fusion protein composed of a tetanus toxoid (TT) T-helper cell epitope and human B7-H4IgV domain (TT-rhB7-H4IgV). We investigated the anti-tumor effect of the TT-rhB7-H4IgV vaccine in BALB/c mice and SP2/0 myeloma growth was significantly suppressed in mice. The TT-rhB7-H4IgV vaccine induced high-titer specific antibodies in mice. Further, the antibodies induced by TT-rhB7-H4IgV vaccine were capable of depleting SP2/0 cells through complement-dependent cytotoxicity (CDC) in vitro. On the other hand, the poor cellular immune response was irrelevant to the therapeutic efficacy. These results indicate that the recombinant TT-rhB7-H4IgV vaccine might be a useful candidate of immunotherapy for the treatment of some tumors associated with abnormal expression of B7-H4. [BMB Reports 2014; 47(7): 399-404]
Collapse
Affiliation(s)
- Nan Mu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Nannan Liu
- Experiment Teaching Center of Basic Medicine, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Yujin Xu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Jialin Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Shouzhen Wu
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, 17 Changle West Road, 710032 Xi'an, Shaanxi, People's Republic of China
| | - Haichuan Su
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, 710038, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
10
|
Zhang C, Wang W, Qin X, Xu Y, Huang T, Hao Q, Li W, Wu S, Zhang Y. B7-H1 protein vaccine induces protective and therapeutic antitumor responses in SP2/0 myeloma-bearing mice. Oncol Rep 2013; 30:2442-8. [PMID: 23970300 DOI: 10.3892/or.2013.2686] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 03/19/2013] [Indexed: 11/06/2022] Open
Abstract
B7-H1 is a co-inhibitory molecule belonging to the B7 family. The B7-H1 protein is only expressed on macrophage lineage of cells in normal tissues, but is overexpressed in most types of tumor. The aberrant expression of cell surface B7-H1 on cancer cells is generally associated with high-risk prognostic factors. The tumor-associated B7-H1 increases apoptosis of antigen-specific T cells through interaction with its receptor PD-1 on CD8+ T cells and contributes to tumor immune evasion. These features suggest that B7-H1 may be a therapeutic target for the B7-H1-expressing tumors. We developed a therapeutic vaccine by coupling a tetanus toxoid T-helper cell epitope with the N-terminal of B7-H1 IgV-like domain. This vaccine was able to induce high titers of antibodies against B7-H1 in mice which were able to bind to native cell surface B7-H1. We chose the B7-H1-expressing SP2/0 myeloma and its syngeneic host (the BALB/c mouse) as the model to study the antitumor activity of the rhB7-H1M vaccine. Vaccination with this modified B7-H1 protein resulted in almost complete protection from SP2/0 tumor challenge and efficiently eliminated pre-established tumors in mice. In addition, B7-H1 vaccination was able to decrease the percentage of CD4+ Foxp3+ regulatory T cells in tumor-bearing mice and which might improve antitumor immunity. These data demonstrate the potential of B7-H1-based vaccine as a therapeutic agent for the treatment of cancer overexpressing B7-H1.
Collapse
Affiliation(s)
- Cun Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Chen YB, Mu CY, Chen C, Huang JA. Association between single nucleotide polymorphism of PD-L1 gene and non-small cell lung cancer susceptibility in a Chinese population. Asia Pac J Clin Oncol 2012; 10:e1-6. [PMID: 23167931 DOI: 10.1111/ajco.12037] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2012] [Indexed: 11/29/2022]
Abstract
AIM To evaluate the correlation between a polymorphism of PD-L1 gene and the susceptibility of non-small cell lung cancer (NSCLC) in a Chinese population. METHODS A total of 293 Chinese patients with NSCLC and 293 age and sex matched controls of the same ethnic origin were enrolled in this study. A/C polymorphism at position 8923 in intron 4 of PD-L1 gene was typed using the polymerase chain reaction-restriction fragment length polymorphism method (PCR-RFLP). The interactions between A/C genotype, allele frequency and NSCLC susceptibility were analyzed. RESULTS The A/C genotype frequencies were significantly different between NSCLC patients and controls. The AC and CC frequencies were higher in NSCLC patients than in controls (16.4 vs 8.9%, 1.0 vs 0.3%, respectively). The C-allele frequency was higher in NSCLC patients than in controls (9.2 vs 4.8%). Significant differences in the A and C allele frequencies were noted between the two groups (χ(2) = 8.864, P = 0.003). More risk of NSCLC was found in individuals carrying the C allele than in those carrying the A allele (OR = 2.203; 95% CI 1.262-3.242). In both light smokers (≤20 pack-years) and heavy smokers (>20 pack-years), individuals carrying the C-allele had more risk of NSCLC than those carrying the A-allele (light smokers OR = 1.847, 95% CI 1.001-3.409; heavy smokers OR = 3.252, 95% CI 1.196-8.845, respectively). CONCLUSION An A/C polymorphism at position 8923 in the PD-L1 gene is associated with NSCLC susceptibility. The PD-L1 polymorphism plays a role in NSCLC, especially in patients with the C-allele.
Collapse
Affiliation(s)
- Yan-bin Chen
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Suzhou, China
| | | | | | | |
Collapse
|
12
|
Abstract
Abstract
The adaptive immune system can be a potent defense mechanism against cancer; however, it is often hampered by immune suppressive mechanisms in the tumor microenvironment. Coinhibitory molecules expressed by tumor cells, immune cells, and stromal cells in the tumor milieu can dominantly attenuate T-cell responses against cancer cells. Today, a variety of coinhibitory molecules, including cytotoxic T lymphocyte–associated antigen-4, programmed death-1, B and T lymphocyte attenuator, LAG3, T-cell immunoglobulin and mucin domain 3, and CD200 receptor, have been implicated in immune escape of cancer cells. Sustained signaling via these coinhibitory molecules results in functional exhaustion of T cells, during which the ability to proliferate, secrete cytokines, and mediate lysis of tumor cells is sequentially lost. In this review, we discuss the influence of coinhibitory pathways in suppressing autologous and allogeneic T cell–mediated immunity against hematologic malignancies. In addition, promising preclinical and clinical data of immunotherapeutic approaches interfering with negative cosignaling, either as monotherapy or in conjunction with vaccination strategies, are reviewed. Numerous studies indicate that coinhibitory signaling hampers the clinical benefit of current immunotherapies. Therefore, manipulation of coinhibitory networks is an attractive adjuvant immunotherapeutic intervention for hematologic cancers after standard treatment with chemotherapy and hematopoietic stem cell transplantation.
Collapse
|
13
|
Norde WJ, Maas F, Hobo W, Korman A, Quigley M, Kester MGD, Hebeda K, Falkenburg JHF, Schaap N, de Witte TM, van der Voort R, Dolstra H. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. Cancer Res 2011; 71:5111-22. [PMID: 21659460 DOI: 10.1158/0008-5472.can-11-0108] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Tumor relapses remain a serious problem after allogeneic stem cell transplantation (alloSCT), despite the long-term persistence of minor histocompatibility antigen (MiHA)-specific memory CD8(+) T cells specific for the tumor. We hypothesized that these memory T cells may lose their function over time in transplanted patients. Here, we offer functional and mechanistic support for this hypothesis, based on immune inhibition by programmed death-1 (PD-1) expressed on MiHA-specific CD8(+) T cells and the associated role of the PD-1 ligand PD-L1 on myeloid leukemia cells, especially under inflammatory conditions. PD-L1 was highly upregulated on immature human leukemic progenitor cells, whereas costimulatory molecules such as CD80 and CD86 were not expressed. Thus, immature leukemic progenitor cells seemed to evade the immune system by inhibiting T-cell function via the PD-1/PD-L1 pathway. Blocking PD-1 signaling using human antibodies led to elevated proliferation and IFN-γ production of MiHA-specific T cells cocultured with PD-L1-expressing leukemia cells. Moreover, patients with relapsed leukemia after initial MiHA-specific T-cell responses displayed high PD-L1 expression on CD34(+) leukemia cells and increased PD-1 levels on MiHA-specific CD8(+) T cells. Importantly, blocking PD-1/PD-L1 interactions augment proliferation of MiHA-specific CD8(+) memory T cells from relapsed patients. Taken together, our findings indicate that the PD-1/PD-L pathway can be hijacked as an immune escape mechanism in hematological malignancies. Furthermore, they suggest that blocking the PD-1 immune checkpoint offers an appealing immunotherapeutic strategy following alloSCT in patients with recurrent or relapsed disease.
Collapse
Affiliation(s)
- Wieger J Norde
- Departments of Laboratory Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Identification of PD-1 as a Unique Marker for Failing Immune Reconstitution in HIV-1–Infected Patients on Treatment. J Acquir Immune Defic Syndr 2011; 56:118-24. [DOI: 10.1097/qai.0b013e3181fbab9f] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|