1
|
de Jong FC, Kvikstad V, Hoedemaeker RF, van der Made ACJ, van der Bosch TP, van Casteren NJ, van Kessel KEM, Zwarthoff EC, Boormans JL, Zuiverloon TCM. PD-L1 expression in high-risk non-muscle invasive bladder cancer is not a biomarker of response to BCG. World J Urol 2025; 43:57. [PMID: 39752014 PMCID: PMC11698758 DOI: 10.1007/s00345-024-05392-5] [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: 01/29/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
PURPOSE Up to 50% of high-risk non-muscle invasive bladder cancer (HR-NMIBC) patients fail Bacillus Calmette-Guérin (BCG) treatment, resulting in a high risk of progression and poor clinical outcomes. Biomarkers that predict outcomes after BCG are lacking. The antitumor effects of BCG are driven by a cytotoxic T cell response, which may be controlled by immune checkpoint proteins like Programmed Death Ligand 1 (PD-L1). Here, we hypothesized that PD-L1 protein expression could serve as a biomarker for BCG-failure. METHODS HR-NMIBC patients who received ≥ 5 BCG instillations were included. Tissue microarrays were constructed from BCG-naïve tumors and recurrences and stained with the PD-L1 (SP142) antibody. PD-L1 status was defined as ≥ 5% tumor-infiltrating immune cells with membrane staining in the tumor area. Clinicopathological associations with PD-L1 positive tumors were investigated, and time-to-event analyses were performed comparing PD-L1 positive vs. negative tumors. RESULTS 432 BCG-naïve tumors and 160 recurrences were included, and 91% of patients received adequate BCG. In BCG-naïve tumors, PD-L1 was expressed in 7% of patients and PD-L1 expression was associated with stage T1 versus Ta disease (p = 0.015). PD-L1 expression was not associated with treatment failure after adequate BCG (p = 0.782) nor with progression-free survival (p = 0.732). Testing cut-offs of ≥ 1% and ≥ 10% PD-L1 positivity did not alter results. High PD-L1 expression was more frequent in tumor recurrences (14%) as compared to BCG-naïve tumors (p = 0.012). CONCLUSION PD-L1 expression in HR-NMIBC is not a biomarker of response to BCG. However, PD-L1 is higher in a subset of tumors that failed BCG treatment. More research is needed to determine the role of PD-L1 in tumors where BCG treatment failed.
Collapse
Affiliation(s)
- Florus C de Jong
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Room Be-304, 3015 GD, Rotterdam, The Netherlands
| | | | | | - Angelique C J van der Made
- Department of Pathology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Niels J van Casteren
- Department of Urology, Franciscus Gasthuis & Vlietland, Rotterdam, The Netherlands
| | | | - Ellen C Zwarthoff
- Department of Pathology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joost L Boormans
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Room Be-304, 3015 GD, Rotterdam, The Netherlands
| | - Tahlita C M Zuiverloon
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, Room Be-304, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Deng X, Liu J. Narrative review on efficacy and safety of anti-angiogenesis in combination with immunotherapy in the treatment of breast cancer. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 5:32. [PMID: 39534580 PMCID: PMC11557168 DOI: 10.21037/tbcr-24-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024]
Abstract
Background and Objective Breast cancer was the second frequently diagnosed cancer in 2022 among all cancers. Besides classical chemotherapy and radiation, immunotherapy and targeted therapy are both identical treatment options for patients with advanced breast cancer. Immunotherapy is a therapeutic approach to control and eliminate tumors by restarting and maintaining the tumor-immunity cycle and restoring the body's normal anti-tumor immune response. Immunotherapy alone or in combination with other therapies has been shown to be clinically beneficial in a variety of solid tumors with a manageable safety profile. However, immunotherapy alone cannot fully satisfy the therapeutic needs for patients with breast cancer. Therefore, there is an urgent need for immunotherapy to be combined with other therapeutic approaches to increase treatment efficacy. Methods We systematically searched PubMed database for relevant studies published over the past 5 years. Articles were screened for eligibility and key data extracted. Key Content and Findings We assess the current breast cancer treatment landscape, summarizing efficacy and safety of recent immunotherapy, chemotherapy combined with immunotherapy, immunotherapy combined with anti-angiogenic therapy. In the treatment of breast cancer, aiming to promote further research and applications of this novel treatment regimen in patients with breast cancer. Since anti-angiogenic therapy can reprogramme the tumor immune microenvironment, immunotherapy in combination with anti-angiogenic therapy might have a synergistic effect, igniting a new hope for immunotherapy for breast cancer patients. The review's conclusions offer insightful information on the state of breast cancer treatment today. In the end, improving clinical practice and pertinent research for immunotherapy combination therapy will contribute to bettering patient outcomes, raising quality of life, and creating more potent treatments. Conclusions This review emphasizes the potential of immunotherapy combinations, especially with anti-angiogenic therapeutic regimens, as a viable strategy for the treatment of breast cancer through a thorough study of the literature. To improve treatment approaches, lessen side effects for patients, and find trustworthy biomarkers to forecast response to immunotherapy combo medicines, further research is necessary.
Collapse
Affiliation(s)
- Xueman Deng
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jieqiong Liu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Nguyen NP, Karlsson UL, Page BR, Chirila ME, Vinh-Hung V, Gorobets O, Arenas M, Mohammadianpanah M, Javadinia SA, Giap H, Kim L, Dutheil F, Murthy V, Mallum AA, Tlili G, Dahbi Z, Loganadane G, Blanco SC, Bose S, Natoli E, Li E, Morganti AG. Immunotherapy and radiotherapy for older patients with invasive bladder cancer unfit for surgery or chemotherapy: practical proposal by the international geriatric radiotherapy group. Front Oncol 2024; 14:1371752. [PMID: 39026981 PMCID: PMC11254657 DOI: 10.3389/fonc.2024.1371752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
The standard of care for non-metastatic muscle invasive bladder cancer is either radical cystectomy or bladder preservation therapy, which consists of maximal transurethral bladder resection of the tumor followed by concurrent chemoradiation with a cisplatin-based regimen. However, for older cancer patients who are too frail for surgical resection or have decreased renal function, radiotherapy alone may offer palliation. Recently, immunotherapy with immune checkpoint inhibitors (ICI) has emerged as a promising treatment when combined with radiotherapy due to the synergy of those two modalities. Transitional carcinoma of the bladder is traditionally a model for immunotherapy with an excellent response to Bacille Calmette-Guerin (BCG) in early disease stages, and with avelumab and atezolizumab for metastatic disease. Thus, we propose an algorithm combining immunotherapy and radiotherapy for older patients with locally advanced muscle-invasive bladder cancer who are not candidates for cisplatin-based chemotherapy and surgery.
Collapse
Affiliation(s)
- Nam Phong Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Ulf Lennart Karlsson
- Department of Radiation Oncology, International Geriatric Radiotherapy Group, Washington, DC, United States
| | - Brandi R. Page
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD, United States
| | - Monica-Emilia Chirila
- Department of Clinical Development, MVision AI, Helsinki, Finland
- Department of Radiation Oncology, Amethyst Radiotherapy Centre, Cluj-Napoca, Romania
| | - Vincent Vinh-Hung
- Department of Radiation Oncology, Centre Hospitalier Public du Contentin, Cherbour-en-Contentin, France
| | - Olena Gorobets
- Department of Oral Surgery, University Hospital of Martinique, Fort-de-France, France
| | - Meritxell Arenas
- Department of Radiation Oncology, Sant Joan de Reus University Hospital, University of Rovira, I Virgili, Tarragona, Spain
| | - Mohammad Mohammadianpanah
- Colorectal Research Center, Department of Radiation Oncology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Alireza Javadinia
- Non-Communicable Diseases Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Huan Giap
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC, United States
| | - Lyndon Kim
- Division of Neuro-Oncology, Mount Sinai Hospital, New York, NY, United States
| | - Fabien Dutheil
- Department of Radiation Oncology, Clinique Sainte Clotilde, Saint Denis, Reunion, France
| | - Vedang Murthy
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Abba Aji Mallum
- Department of Radiation Oncology, University of KwaZulu Natal, Durban, South Africa
| | - Ghassen Tlili
- Department of Urology, University Hospital Center, Sousse, Tunisia
| | - Zineb Dahbi
- Department of Radiation Oncology, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | | | - Sergio Calleja Blanco
- Department of Oral Maxillofacial Surgery, Howard University, Washington, DC, United States
| | - Satya Bose
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Elena Natoli
- Department of Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Radiation Oncology, DIMEC, Bologna University, Bologna, Italy
| | - Eric Li
- Department of Pathology, Howard University, Washington, DC, United States
| | - Alessio G. Morganti
- Department of Radiation Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Radiation Oncology, DIMEC, Bologna University, Bologna, Italy
| |
Collapse
|
4
|
Bedke J, Black PC, Szabados B, Guerrero-Ramos F, Shariat SF, Xylinas E, Brinkmann J, Blake-Haskins JA, Cesari R, Redorta JP. Optimizing outcomes for high-risk, non-muscle-invasive bladder cancer: The evolving role of PD-(L)1 inhibition. Urol Oncol 2023; 41:461-475. [PMID: 37968169 DOI: 10.1016/j.urolonc.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Transurethral resection of bladder tumor followed by intravesical Bacillus Calmette-Guérin (BCG) is the standard of care in high-risk, non-muscle-invasive bladder cancer (NMIBC). Although many patients respond, recurrence and progression are common. In addition, patients may be unable to receive induction + maintenance due to intolerance or supply issues. Therefore, alternative treatment options are urgently required. Programmed cell death (ligand) 1 (PD-[L]1) inhibitors show clinical benefit in phase 1/2 trials in BCG-unresponsive NMIBC patients. This review presents the status of PD-(L)1 inhibition in high-risk NMIBC and discusses future directions. PubMed and Google scholar were searched for articles relating to NMIBC immunotherapy and ClinicalTrials.gov for planned and ongoing clinical trials. Preclinical and early clinical studies show that BCG upregulates PD-L1 expression in bladder cancer cells and, when combined with a PD-(L)1 inhibitor, a potent antitumor response is activated. Based on this mechanism, several PD-(L)1 inhibitors are in phase 3 trials in BCG-naïve, high-risk NMIBC in combination with BCG. Whereas PD-(L)1 inhibitors are well characterized in patients with advanced malignancies, the impact of immune-related adverse events (irAE) on the benefit/risk ratio in NMIBC should be determined. Alternative routes to intravenous administration, like subcutaneous and intravesical administration, may facilitate adherence and access. The outcomes of combination of PD-(L)1 inhibitors and BCG in NMIBC are highly anticipated. There will be a need to address treatment resources, optimal management of irAEs and education and training related to use of this therapy in clinical practice.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology and Transplantation Surgery, Kilinikum Stuttgart, Stuttgart, Germany.
| | - Peter C Black
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
| | - Bernadett Szabados
- University College London Hospital, London, UK; Bart's Cancer Institute, Queen Mary University of London, London, UK
| | | | | | - Evanguelos Xylinas
- Department of Urology, Hôpital Bichat - Claude-Bernard, Université de Paris Cité, Paris, France
| | | | | | | | | |
Collapse
|
5
|
Sampaio-Ribeiro G, Ruivo A, Silva A, Santos AL, Oliveira RC, Gama J, Cipriano MA, Tralhão JG, Paiva A. Innate Immune Cells in the Tumor Microenvironment of Liver Metastasis from Colorectal Cancer: Contribution to a Comprehensive Therapy. Cancers (Basel) 2023; 15:3222. [PMID: 37370832 DOI: 10.3390/cancers15123222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent type of cancer, and liver metastasis is the most common site of metastatic development. In the tumor microenvironment (TME), various innate immune cells are known to influence cancer progression and metastasis occurrence. CD274 (PD-L1) and CD206 (MRC1) are proteins that have been associated with poor prognosis and disease progression. We conducted a study on tumoral and non-tumoral biopsies from 47 patients with CRC liver metastasis, using flow cytometry to phenotypically characterize innate immune cells. Our findings showed an increase in the expression of CD274 on classical, intermediate, and non-classical monocytes when comparing tumor with non-tumor samples. Furthermore, tumor samples with a desmoplastic growth pattern exhibited a significantly decreased percentage of CD274- and CD206-positive cells in all monocyte populations compared to non-desmoplastic samples. We found a correlation between a lower expression of CD206 or CD274 on classical, intermediate, and non-classical monocytes and increased disease-free survival, which points to a better prognosis for these patients. In conclusion, our study has identified potential new targets and biomarkers that could be incorporated into a personalized medicine approach to enhance the outcome for colorectal cancer patients.
Collapse
Affiliation(s)
- Gabriela Sampaio-Ribeiro
- Flow Cytometry Unit, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra EPE, 3000-075 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Ruivo
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Silva
- Flow Cytometry Unit, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra EPE, 3000-075 Coimbra, Portugal
| | - Ana Lúcia Santos
- Flow Cytometry Unit, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra EPE, 3000-075 Coimbra, Portugal
| | - Rui Caetano Oliveira
- Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Germano de Sousa-Centro de Diagnóstico Histopatológico CEDAP, 3000-377 Coimbra, Portugal
- Centre of Investigation on Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - João Gama
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - Maria Augusta Cipriano
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
| | - José Guilherme Tralhão
- Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Surgery Department, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Centre of Investigation on Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Center of Coimbra (CACC), 3000-075 Coimbra, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra EPE, 3000-075 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, ESTESC-Coimbra Health School, Instituto Politécnico de Coimbra, 3046-854 Coimbra, Portugal
| |
Collapse
|
6
|
Lui G, Minnar CM, Soon-Shiong P, Schlom J, Gameiro SR. Exploiting an Interleukin-15 Heterodimeric Agonist (N803) for Effective Immunotherapy of Solid Malignancies. Cells 2023; 12:1611. [PMID: 37371081 DOI: 10.3390/cells12121611] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023] Open
Abstract
Identifying effective immunotherapies for solid tumors remains challenging despite the significant clinical responses observed in subsets of patients treated with immune checkpoint inhibitors. Interleukin-15 (IL-15) is a promising cytokine for the treatment of cancer as it stimulates NK and CD8+ lymphocytes. However, unfavorable pharmacokinetics and safety concerns render recombinant IL-15 (rIL-15) a less attractive modality. These shortcomings were addressed by the clinical development of heterodimeric IL-15 agonists, including N803. In preclinical tumor models, N803 elicited significant Th1 immune activation and tumor suppressive effects, primarily mediated by NK and CD8+ T lymphocytes. In addition, multiple clinical studies have demonstrated N803 to be safe for the treatment of cancer patients. The combination of N803 with the immune checkpoint inhibitor nivolumab demonstrated encouraging clinical responses in nivolumab-naïve and nivolumab-refractory patients with non-small cell lung cancer. In a recent Phase II/III clinical study, most Bacillus Calmette-Guerin (BCG)-refractory bladder cancer patients treated with N803 plus BCG experienced durable complete responses. Currently, N803 is being evaluated preclinically and clinically in combination with various agents, including chemotherapeutics, immune checkpoint inhibitors, vaccines, and other immuno-oncology agents. This report will review the mechanism(s) of action of N803 and how it relates to the preclinical and clinical studies of N803.
Collapse
Affiliation(s)
- Grace Lui
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Christine M Minnar
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sofia R Gameiro
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Tumor-Microenvironment Characterization of the MB49 Non-Muscle-Invasive Bladder-Cancer Orthotopic Model towards New Therapeutic Strategies. Int J Mol Sci 2022; 24:ijms24010123. [PMID: 36613562 PMCID: PMC9820528 DOI: 10.3390/ijms24010123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Bacillus Calmette-Guérin (BCG) instillations for the treatment of non-muscle-invasive bladder cancer patients can result in significant side effects and treatment failure. Immune checkpoint blockade and/or decreasing tumor-infiltrating myeloid suppressor cells may be alternative or complementary treatments. Here, we have characterized immune cell infiltration and chemoattractant molecules in mouse orthotopic MB49 bladder tumors. Our data show a 100-fold increase in CD45+ immune cells from day 5 to day 9 tumors including T cells and mainly myeloid cells. Both monocytic myeloid-derived suppressor-cells (M-MDSC) and polymorphonuclear (PMN)-MDSC were strongly increased in day 9 tumors, with PMN-MDSC representing ca. 70% of the myeloid cells in day 12 tumors, while tumor associated macrophages (TAM) were only modestly increased. The kinetic of PD-L1 tumor expression correlated with published data from patients with PD-L1 expressing bladder tumors and with efficacy of anti-PD-1 treatment, further validating the orthotopic MB49 bladder-tumor model as suitable for designing novel therapeutic strategies. Comparison of chemoattractants expression during MB49 bladder tumors grow highlighted CCL8 and CCL12 (CCR2-ligands), CCL9 and CCL6 (CCR-1-ligands), CXCL2 and CXCL5 (CXCR2-ligands), CXCL12 (CXCR4-ligand) and antagonist of C5/C5a as potential targets to decrease myeloid suppressive cells. Data obtained with a single CCR2 inhibitor however showed that the complex chemokine crosstalk would require targeting multiple chemokines for anti-tumor efficacy.
Collapse
|
8
|
Ren Y, Song J, Li X, Luo N. Rationale and Clinical Research Progress on PD-1/PD-L1-Based Immunotherapy for Metastatic Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23168878. [PMID: 36012144 PMCID: PMC9408844 DOI: 10.3390/ijms23168878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/26/2022] [Accepted: 08/04/2022] [Indexed: 01/23/2023] Open
Abstract
Metastatic triple-negative breast cancer (mTNBC), a highly aggressive and malignant tumor, currently lacks an effective treatment. There has been some progress in the treatment of mTNBC with programmed death receptor-1/programmed death ligand-1 (PD-1/PD-L1) immunotherapy in recent years. The combination of PD-1/PD-L1 inhibitors with other therapies is a noteworthy treatment strategy. Immunotherapy in combination with chemotherapy or small-molecule inhibitors still faces many challenges. Additionally, there are some new immunotherapy targets in development. We aimed to further evaluate the effectiveness and usefulness of immunotherapy for treating mTNBC and to propose new immunotherapy strategies. This review explains the rationale and results of existing clinical trials evaluating PD-1/PD-L1 inhibitors alone or in combination for the treatment of mTNBC. For patients with aggressive tumors and poor health, PD-1/PD-L1 inhibitors, either alone or in combination with other modalities, have proven to be effective. However, more research is needed to explore more effective immunotherapy regimens that will lead to new breakthroughs in the treatment of mTNBC.
Collapse
|
9
|
Tsuji S, Reil K, Nelson K, Proclivo VH, McGuire KL, Giacalone MJ. Intravesical VAX014 Synergizes with PD-L1 Blockade to Enhance Local and Systemic Control of Bladder Cancer. Cancer Immunol Res 2022; 10:978-995. [PMID: 35679299 DOI: 10.1158/2326-6066.cir-21-0879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/08/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022]
Abstract
Emerging clinical evidence indicates that the combination of local administration of immunotherapy with systemic immune checkpoint blockade targeting the PD-1/PD-L1 pathway improves response rates in select solid tumor indications; however, limited clinical experience with this approach exists in advanced bladder cancer patients. VAX014 is a novel bacterial minicell-based, integrin-targeted oncolytic agent undergoing clinical investigation for intravesical (IVE) treatment of non-muscle invasive bladder cancer. Here, we demonstrated that the antitumor activity of VAX014 following IVE administration was dependent upon CD4+ and CD8+ T cells in two syngeneic orthotopic bladder tumor models (MB49 and MBT-2). PD-L1 upregulation was found to be an acquired immune-resistance mechanism in the MB49 model, and the combination of VAX014 with systemic PD-L1 blockade resulted in a significant improvement in bladder tumor clearance rates and development of protective antitumor immunologic memory. Combination treatment also led to enhanced systemic antitumor immune responses capable of clearing distal intradermal tumors and controlling pulmonary metastasis. Distal tumors actively responding to combination therapy demonstrated a phenotypic shift from Treg to Th1 in intratumoral CD4+ T cells, which was accompanied by a higher percentage of activated CD8+ T cells and higher IFNγ. Finally, VAX014's target integrins α3β1 and α5β1 were overexpressed in tumor biopsies from advanced stage bladder cancer patients, as well as in both the MB49 and MBT-2 orthotopic mouse models of bladder cancer. These collective findings provide rationale for clinical investigation of VAX014 and systemic PD-1/PD-L1 blockade in advanced stage bladder cancer.
Collapse
Affiliation(s)
- Shingo Tsuji
- Vaxiion Therapeutics (United States), San Diego, United States
| | - Katherine Reil
- Vaxiion Therapeutics and San Diego State University, San Diego, United States
| | - Kinsey Nelson
- Vaxiion Therapeutics and San Diego State University, San Diego, CA, United States
| | | | | | | |
Collapse
|
10
|
Huang KC, Chanda D, McGrath S, Dixit V, Zhang C, Wu J, Tendyke K, Yao H, Hukkanen R, Taylor N, Verbel D, Kim DS, Endo A, Noland TA, Chen Y, Matijevic M, Wang J, Hutz J, Sarwar N, Fang FG, Bao X. Pharmacological Activation of STING in Bladder Induces Potent Anti-tumor Immunity in Non-Muscle Invasive Murine Bladder Cancer. Mol Cancer Ther 2022; 21:914-924. [PMID: 35313332 DOI: 10.1158/1535-7163.mct-21-0780] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/25/2022] [Accepted: 03/09/2022] [Indexed: 11/16/2022]
Abstract
Schweinfurthins, a class of natural products, have attracted considerable interest for novel therapy development because of their selective and potent anti-proliferative activity against human cancer cells. However, the underlying mechanism is not well understood. Herein, we demonstrated that schweinfurthins preferentially inhibited the proliferation of PTEN deficient cancer cells by indirect inhibition of AKT phosphorylation. Intracellularly, schweinfurthins and their analogs arrested trans-Golgi-network trafficking, likely by binding to oxysterol-binding proteins, leading to an effective inhibition of mTOR/AKT signaling through inducing endoplasmic reticulum stress and suppressing both lipid raft-mediated PI3K activation and mTOR/RheB complex formation. Moreover, schweinfurthins were found to be highly potent toward PTEN-deficient B cell lymphoma cells, and displayed two orders of magnitude lower activity toward normal human peripheral blood mononuclear cells and primary fibroblasts in vitro. These results revealed a previously unrecognized role of schweinfurthins in trans-Golgi-network trafficking and linked mechanistically this cellular effect with mTOR/AKT signaling and with cancer cell survival and growth. Our findings suggest a new opportunity to modulate oncogenic signaling by interfering with TGN trafficking to treat mTOR/AKT-dependent human cancers.
Collapse
Affiliation(s)
| | - Dinesh Chanda
- Johnson & Johnson (United States), Cambridge, MA, United States
| | | | | | - Chi Zhang
- Dewpoint Therapeutics, Boston, United States
| | - Jiayi Wu
- H3biomedicine Inc, United States
| | | | - Huilan Yao
- H3 Biomedicine, Cambridge, United States
| | | | - Noel Taylor
- Eisai (United States), Andover, United States
| | | | | | | | | | - Yu Chen
- Eisai Inc, Cambridge, United States
| | - Mark Matijevic
- Boston Pharmaceuticals, Cambridge, Massachusetts, United States
| | - John Wang
- Eisai (United States), Andover, Massachusetts, United States
| | - Janna Hutz
- Eisai, Inc., Cambridge, MA, United States
| | | | | | - Xingfeng Bao
- H3biomedicine Inc., Cambridge, MA, United States
| |
Collapse
|
11
|
Terranova N, French J, Dai H, Wiens M, Khandelwal A, Ruiz‐Garcia A, Manitz J, Heydebreck A, Ruisi M, Chin K, Girard P, Venkatakrishnan K. Pharmacometric modeling and machine learning analyses of prognostic and predictive factors in the JAVELIN Gastric 100 phase III trial of avelumab. CPT Pharmacometrics Syst Pharmacol 2022; 11:333-347. [PMID: 34971492 PMCID: PMC8923733 DOI: 10.1002/psp4.12754] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/23/2022] Open
Abstract
Avelumab (anti–PD‐L1) is an approved anticancer treatment for several indications. The JAVELIN Gastric 100 phase III trial did not meet its primary objective of demonstrating superior overall survival (OS) with avelumab maintenance versus continued chemotherapy in patients with advanced gastric cancer/gastroesophageal junction cancer; however, the OS rate was numerically higher with avelumab at timepoints after 12 months. Machine learning (random forests, SIDEScreen, and variable‐importance assessments) was used to build models to identify prognostic/predictive factors associated with long‐term OS and tumor growth dynamics (TGDs). Baseline, re‐baseline, and longitudinal variables were evaluated as covariates in a parametric time‐to‐event model for OS and Gompertzian population model for TGD. The final OS model incorporated a treatment effect on the log‐logistic shape parameter but did not identify a treatment effect on OS or TGD. Variables identified as prognostic for longer OS included older age; higher gamma‐glutamyl transferase (GGT) or albumin; absence of peritoneal carcinomatosis; lower neutrophil‐lymphocyte ratio, lactate dehydrogenase, or C‐reactive protein (CRP); response to induction chemotherapy; and Eastern Cooperative Oncology Group performance status of 0. Among baseline and time‐varying covariates, the largest effects were found for GGT and CRP, respectively. Liver metastasis at re‐baseline predicted higher tumor growth. Tumor size after induction chemotherapy was associated with number of metastatic sites and stable disease (vs. response). Asian region did not impact OS or TGD. Overall, an innovative workflow supporting pharmacometric modeling of OS and TGD was established. Consistent with the primary trial analysis, no treatment effect was identified. However, potential prognostic factors were identified.
Collapse
Affiliation(s)
- Nadia Terranova
- Merck Institute of Pharmacometrics (an affiliate of Merck KGaA, Darmstadt, Germany) Lausanne Switzerland
| | | | | | | | | | | | | | | | | | | | - Pascal Girard
- Merck Institute of Pharmacometrics (an affiliate of Merck KGaA, Darmstadt, Germany) Lausanne Switzerland
| | | |
Collapse
|
12
|
Hrinczenko B, Iannotti N, Goel S, Spigel D, Safran H, Taylor MH, Bennouna J, Wong DJ, Kelly K, Verschraegen C, Bajars M, Manitz J, Ruisi M, Gulley JL. Long-term avelumab in advanced non-small-cell lung cancer: summaries and post hoc analyses from JAVELIN Solid Tumor. Future Oncol 2022; 18:1333-1342. [PMID: 35144482 PMCID: PMC9066292 DOI: 10.2217/fon-2021-0930] [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] [Indexed: 11/21/2022] Open
Abstract
Background: This study examined patients with advanced non-small-cell lung cancer who received long-term avelumab (anti-PD-L1) in a large phase Ib trial (JAVELIN Solid Tumor). Methods: Patients receiving >2 years of avelumab were reviewed and exploratory descriptive analyses were conducted. Results: Individuals with varying baseline characteristics who had received up to 6 years of avelumab were reviewed. Overall, 37/340 (10.9%) had received ≥2 years of treatment; in this subgroup, best response was complete response in 5.4%, partial response in 59.5% and stable disease in 29.7%; 51.4% had continued treatment beyond disease progression. Conclusions: In this study, 11% of patients with advanced non-small-cell lung cancer received ≥2 years of avelumab treatment and experienced prolonged response or continued clinical benefit. Clinical Trial Registration:NCT02395172 (ClinicalTrials.gov)
Collapse
Affiliation(s)
- Borys Hrinczenko
- Division of Hematology/Oncology, Michigan State University, East Lansing, MI 48824, USA
| | - Nicholas Iannotti
- Hematology Oncology Associates of The Treasure Coast, Port St Lucie, FL 34952, USA
| | - Sanjay Goel
- Department of Medical Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Spigel
- Sarah Cannon Research Institute, Nashville, TN 37203, USA
| | - Howard Safran
- Life Span Cancer Institute, Providence, RI 02903, USA
| | - Matthew H Taylor
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jaafar Bennouna
- Department of Pneumology, Thoracic Oncology Unit, Université Hospital of Nantes, Nantes, France
| | - Deborah J Wong
- Los Angeles Medical Center, University of California, Los Angeles, CA 90095, USA
| | - Karen Kelly
- University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Claire Verschraegen
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43221, USA
| | | | - Juliane Manitz
- EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Mary Ruisi
- EMD Serono Research & Development Institute, Inc., Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - James L Gulley
- Genitourinary Malignancies Branch & Laboratory of Tumor Immunology & Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Mehany ABM, Belal A, Mohamed AF, Shaaban S, Abdelhamid G. Apoptotic and anti-angiogenic effects of propolis against human bladder cancer: molecular docking and in vitro screening. Biomarkers 2022; 27:138-150. [PMID: 34927500 DOI: 10.1080/1354750x.2021.2020903] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Bladder cancer is still of unknown initiation and progression, it is difficult to treat the patient once bladder cancer have a distant metastasis. MATERIALS AND METHODS In the present study, propolis extract was evaluated against bladder cancer cells (T24). Two independent pathways were investigated, apoptosis and angiogenesis, Bax, Bcl-2, P53, and caspase-3 for apoptosis, vascular endothelial growth factor receptor and protein kinase A as angiogenesis potential targets. OBJECTIVES Molecular docking studies will be conducted for the major known constituents of Egyptian propolis into apoptotic and angiogenic protein targets, to give better insights to the possible binding mode and interactions and investigate the ability of propolis constituents to target both apoptotic and angiogenic pathways. RESULTS Propolis showed anti-proliferative activity against T24 cancer cell line, the IC50 value was 6.36 µg/ml. Also significant effects of propolis on Bax, Bcl-2, P53, and caspase-3 were observed. DISCUSSION These obtained results proved the ability of propolis to induce cell death. Also it has revealed noticeable effects on protein kinase A and vascular endothelial growth factor receptor. CONCLUSION The obtained results can encourage us to say that propolis extract can induce a programmed cell death in human bladder cancer cells, and also affect angiogenesis.
Collapse
Affiliation(s)
- Ahmed B M Mehany
- Genetic Engineering, Department of Zoology, Faculty of Science Al-Azhar University, Cairo, Egypt
| | - Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aly Fahmy Mohamed
- Holding Company for Production of Vaccines and Biological Products (VACSERA), Agouza, Egypt
| | - Salwa Shaaban
- Department of Microbiology& Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.,Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Ghada Abdelhamid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Egypt
| |
Collapse
|
14
|
Zhou Q, Ding W, Qian Z, Zhu Q, Sun C, Yu Q, Tai Z, Xu K. Immunotherapy Strategy Targeting Programmed Cell Death Ligand 1 and CD73 with Macrophage-Derived Mimetic Nanovesicles to Treat Bladder Cancer. Mol Pharm 2021; 18:4015-4028. [PMID: 34648293 DOI: 10.1021/acs.molpharmaceut.1c00448] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Combination immunotherapy is a promising strategy to remove the inhibitory effect of the tumor microenvironment on immune effector cells, improving the efficacy of immune checkpoint inhibitor treatment in bladder cancer. However, it is challenging to deliver multiple drugs to the tumor tissue effectively and simultaneously to ensure optimal therapeutic effects. Macrophage-derived exosome-mimetic nanovesicles (EMVs) were designed and validated as a nanoplatform for coloading and delivery of the CD73 inhibitor (AB680) and the monoclonal antibody to programmed cell death ligand 1 (aPDL1). The tumor-targeting, biosafety, and therapeutic effects of these nanocomplexes (AB680@EMVs-aPDL1), as a combined immunotherapy strategy for bladder cancer, were assessed in vitro and in vivo. Our results indicate that the nanodrug system was highly stable, provided adequate biosafety, and enhanced tumor targeting in a mouse model of bladder cancer. Moreover, the CD73 inhibitor reduced extracellular adenosine production, and the combination therapy significantly promoted the activation and infiltration of cytotoxic T-lymphocytes, which helped to optimally suppress tumor growth and extend median survival in vivo. Therefore, using EMVs to deliver a combination of aPDL1 and the CD73 inhibitor may be a useful combined immunotherapy strategy for treating bladder cancer.
Collapse
Affiliation(s)
- Qidong Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhiyu Qian
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Quangang Zhu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qin Yu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Zongguang Tai
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
15
|
Wang B, He Z, Yu H, Ou Z, Chen J, Yang M, Fan X, Lin T, Huang J. Intravesical Pseudomonas aeruginosa mannose-sensitive Hemagglutinin vaccine triggers a tumor-preventing immune environment in an orthotopic mouse bladder cancer model. Cancer Immunol Immunother 2021; 71:1507-1517. [PMID: 34718847 DOI: 10.1007/s00262-021-03063-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 09/16/2021] [Indexed: 02/02/2023]
Abstract
Bacillus Calmette-Guerin (BCG) immunotherapy can prevent recurrence and progression in selected patients with non-muscle-invasive bladder cancer (NMIBC); however, significant adverse events and treatment failure suggest the need for alternative agents. A commercial anti-infection vaccine comprises a genetically engineered heat-killed Pseudomonas aeruginosa (PA) expressing many mannose-sensitive hemagglutination (MSHA) fimbriae, termed PA-MSHA, which could be a candidate for bladder cancer intravesical therapy. In an immunocompetent orthotopic MB49 bladder cancer model, we characterized the antitumor effects and mechanisms of PA-MSHA compared with those of BCG. Three weekly intravesical PA-MSHA or BCG treatments reduced tumor involvement; however, only PA-MSHA prolonged survival against MB49 implantation significantly. In non-tumor-bearing mice after treatment, flow-cytometry analysis showed PA-MSHA and BCG induced an increased CD4/CD8 ratio, the levels of effector memory T cell phenotypes (CD44, CXCR-3, and IFN-γ), and the proportion of CD11b+Ly6G-Ly6C-IA/IE+ mature macrophages, but a decrease in the proportion of CD11b+Ly6G-Ly6C+IA/IE- monocytic myeloid-derived suppressor cells (Mo-MDSCs) and the expression of suppressive molecules on immune cells (PD-L1, PD-1, TIM-3, and LAG-3). Notably, PA-MSHA, but not BCG, significantly reduced PD-1 and TIM-3 expression on CD4+ T cells, which might account for the better effects of PA-MSHA than BCG. However, in tumor-bearing mice after treatment, the increased proportion of Mo-MDSCs and high expression of PD-L1 might be involved in treatment failure. Thus, modulating the balance among adaptive and innate immune responses was identified as a key process underlying PA-MSHA-mediated treatment efficacy. The results demonstrated mechanisms underlying intravesical PA-MSHA therapy, pointing at its potential as an alternative effective treatment for NMIBC.
Collapse
Affiliation(s)
- Bo Wang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Zhihua He
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China.,Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Hao Yu
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China
| | - Ziwei Ou
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China
| | - Junyu Chen
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China
| | - Meihua Yang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China
| | - Xinxiang Fan
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China
| | - Tianxin Lin
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| | - Jian Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen (Zhongshan) University, Guangzhou, 510120, People's Republic of China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
16
|
Rijnders M, Balcioglu HE, Robbrecht DGJ, Oostvogels AAM, Wijers R, Aarts MJB, Hamberg P, van Leenders GJLH, Nakauma-González JA, Voortman J, Westgeest HM, Boormans JL, de Wit R, Lolkema MP, van der Veldt AAM, Debets R. Anti-PD-1 Efficacy in Patients with Metastatic Urothelial Cancer Associates with Intratumoral Juxtaposition of T Helper-Type 1 and CD8 + T cells. Clin Cancer Res 2021; 28:215-226. [PMID: 34615720 DOI: 10.1158/1078-0432.ccr-20-3319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 04/08/2021] [Accepted: 09/29/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE PD-1 inhibition results in durable antitumor responses in a proportion of patients with metastatic urothelial cancer (mUC). The majority of patients, however, do not experience clinical benefit. In this study, we aimed to identify early changes in T-cell subsets that underlie anti-PD-1 efficacy in patients with mUC. EXPERIMENTAL DESIGN Paired samples were collected from peripheral blood, plasma, and metastatic lesions of 56 patients with mUC at baseline and weeks 6 and 12 after initiating pembrolizumab treatment (200 mg intravenously, every 3 weeks). Samples were analyzed using multiplex flow cytometry, ELISA, and in situ stainings, including cellular network analysis. Treatment response was evaluated as best overall response according to RECIST v1.1, and patients were classified as responder (complete or partial response) or nonresponder (progressive disease). RESULTS In responders, baseline fractions of CD4+ T cells expressing cosignaling receptors were higher compared with nonresponders. The fraction of circulating PD-1+ CD4+ T cells decreased at weeks 6 and 12, whereas the fraction of 4-1BB+ CD28+ CD4+ T cells increased at week 12. In metastatic lesions of responders, the baseline density of T helper-type 1 (Th1) cells, defined as T-bet+ CD4+ T cells, was higher as compared to non-responders. Upon treatment, Th1 cells became localized in close proximity to CD8+ T cells, CD11b+ myeloid cells, and tumor cells. CONCLUSIONS A decrease in the fraction of circulating PD-1+ CD4+ T cells, and juxtaposition of Th1, CD8+, and myeloid cells was associated with response to anti-PD-1 treatment in patients with mUC.
Collapse
Affiliation(s)
- Maud Rijnders
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Hayri E Balcioglu
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Debbie G J Robbrecht
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Astrid A M Oostvogels
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Rebecca Wijers
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Maureen J B Aarts
- Department of Medical Oncology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Paul Hamberg
- Department of Medical Oncology, Franciscus Gasthuis & Vlietland Hospital, Rotterdam, the Netherlands
| | - Geert J L H van Leenders
- Department of Pathology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - J Alberto Nakauma-González
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.,Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.,Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Jens Voortman
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, the Netherlands
| | - Hans M Westgeest
- Department of Medical Oncology, Amphia Hospital Breda, Breda, the Netherlands
| | - Joost L Boormans
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Ronald de Wit
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.,Department of Radiology & Nuclear Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands
| | - Reno Debets
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
17
|
Zhou Q, Qian Z, Ding W, Jiang G, Sun C, Xu K. Chronic Psychological Stress Attenuates the Efficacy of anti-PD-L1 Immunotherapy for Bladder Cancer in Immunocompetent Mice. Cancer Invest 2021; 39:571-581. [PMID: 34148483 DOI: 10.1080/07357907.2021.1943746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We aimed to explore whether chronic psychological stress affects the efficacy of immune checkpoint inhibitors (ICIs) immunotherapy in bladder cancer. The chronic unpredictable mild stress (CUMS) process was applied during the administration of anti-PD-L1 for subcutaneous tumors in mice. Tumor regression was obviously shown in anti-PD-L1 therapy groups, while this effect was notably attenuated by CUMS. Additionally, increased infiltration of regulatory T-cells, decreased amount of CD8+ lymphocytes, and reduced levels of tumor-associated cytokines in tumor sites were observed in mice treated with anti-PD-L1 under CUMS. Therefore, chronic psychological stress could weaken the potency of anti-PD-L1 immunotherapy for bladder cancer.
Collapse
Affiliation(s)
- Qidong Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhiyu Qian
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Weihong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Guangliang Jiang
- Department of Urology, Ruijin Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Chuanyu Sun
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ke Xu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Discovery of Novel Small-Molecule Inhibitors of PD-1/PD-L1 Interaction via Structural Simplification Strategy. Molecules 2021; 26:molecules26113347. [PMID: 34199417 PMCID: PMC8199613 DOI: 10.3390/molecules26113347] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Blockade of the programmed cell death 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) interaction is currently the focus in the field of cancer immunotherapy, and so far, several monoclonal antibodies (mAbs) have achieved encouraging outcomes in cancer treatment. Despite this achievement, mAbs-based therapies are struggling with limitations including poor tissue and tumor penetration, long half-life time, poor oral bioavailability, and expensive production costs, which prompted a shift towards the development of the small-molecule inhibitors of PD-1/PD-L1 pathways. Even though many small-molecule inhibitors targeting PD-1/PD-L1 interaction have been reported, their development lags behind the corresponding mAb, partly due to the challenges of developing drug-like small molecules. Herein, we report the discovery of a series of novel inhibitors targeting PD-1/PD-L1 interaction via structural simplification strategy by using BMS-1058 as a starting point. Among them, compound A9 stands out as the most promising candidate with excellent PD-L1 inhibitory activity (IC50 = 0.93 nM, LE = 0.43) and high binding affinity to hPD-L1 (KD = 3.64 nM, LE = 0.40). Furthermore, A9 can significantly promote the production of IFN-γ in a dose-dependent manner by rescuing PD-L1 mediated T-cell inhibition in Hep3B/OS-8/hPD-L1 and CD3-positive T cells co-culture assay. Taken together, these results suggest that A9 is a promising inhibitor of PD-1/PD-L1 interaction and is worthy for further study.
Collapse
|
19
|
Pramil E, Dillard C, Escargueil AE. Colorectal Cancer and Immunity: From the Wet Lab to Individuals. Cancers (Basel) 2021; 13:cancers13071713. [PMID: 33916641 PMCID: PMC8038567 DOI: 10.3390/cancers13071713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Tackling the current dilemma of colorectal cancer resistance to immunotherapy is puzzling and requires novel therapeutic strategies to emerge. However, characterizing the intricate interactions between cancer and immune cells remains difficult because of the complexity and heterogeneity of both compartments. Developing rationales is intellectually feasible but testing them can be experimentally challenging and requires the development of innovative procedures and protocols. In this review, we delineated useful in vitro and in vivo models used for research in the field of immunotherapy that are or could be applied to colorectal cancer management and lead to major breakthroughs in the coming years. Abstract Immunotherapy is a very promising field of research and application for treating cancers, in particular for those that are resistant to chemotherapeutics. Immunotherapy aims at enhancing immune cell activation to increase tumor cells recognition and killing. However, some specific cancer types, such as colorectal cancer (CRC), are less responsive than others to the current immunotherapies. Intrinsic resistance can be mediated by the development of an immuno-suppressive environment in CRC. The mutational status of cancer cells also plays a role in this process. CRC can indeed be distinguished in two main subtypes. Microsatellite instable (MSI) tumors show a hyper-mutable phenotype caused by the deficiency of the DNA mismatch repair machinery (MMR) while microsatellite stable (MSS) tumors show a comparatively more “stable” mutational phenotype. Several studies demonstrated that MSI CRC generally display good prognoses for patients and immunotherapy is considered as a therapeutic option for this type of tumors. On the contrary, MSS metastatic CRC usually presents a worse prognosis and is not responsive to immunotherapy. According to this, developing new and innovative models for studying CRC response towards immune targeted therapies has become essential in the last years. Herein, we review the in vitro and in vivo models used for research in the field of immunotherapy applied to colorectal cancer.
Collapse
Affiliation(s)
- Elodie Pramil
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Alliance Pour la Recherche en Cancérologie—APREC, Tenon Hospital, F-75012 Paris, France
| | - Clémentine Dillard
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Alliance Pour la Recherche en Cancérologie—APREC, Tenon Hospital, F-75012 Paris, France
| | - Alexandre E. Escargueil
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, F-75012 Paris, France; (E.P.); (C.D.)
- Correspondence: ; Tel.: +33-(0)1-49-28-46-44
| |
Collapse
|
20
|
Reyes RM, Deng Y, Zhang D, Ji N, Mukherjee N, Wheeler K, Gupta HB, Padron AS, Kancharla A, Zhang C, Garcia M, Kornepati AVR, Boyman O, Conejo-Garcia JR, Svatek RS, Curiel TJ. CD122-directed interleukin-2 treatment mechanisms in bladder cancer differ from αPD-L1 and include tissue-selective γδ T cell activation. J Immunother Cancer 2021; 9:e002051. [PMID: 33849925 PMCID: PMC8051418 DOI: 10.1136/jitc-2020-002051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Anti-programmed death-ligand 1 (αPD-L1) immunotherapy is approved to treat bladder cancer (BC) but is effective in <30% of patients. Interleukin (IL)-2/αIL-2 complexes (IL-2c) that preferentially target IL-2 receptor β (CD122) augment CD8+ antitumor T cells known to improve αPD-L1 efficacy. We hypothesized that the tumor microenvironment, including local immune cells in primary versus metastatic BC, differentially affects immunotherapy responses and that IL-2c effects could differ from, and thus complement αPD-L1. METHODS We studied mechanisms of IL-2c and αPD-L1 efficacy using PD-L1+ mouse BC cell lines MB49 and MBT-2 in orthotopic (bladder) and metastatic (lung) sites. RESULTS IL-2c reduced orthotopic tumor burden and extended survival in MB49 and MBT-2 BC models, similar to αPD-L1. Using antibody-mediated cell depletions and genetically T cell-deficient mice, we unexpectedly found that CD8+ T cells were not necessary for IL-2c efficacy against tumors in bladder, whereas γδ T cells, not reported to contribute to αPD-L1 efficacy, were indispensable for IL-2c efficacy there. αPD-L1 responsiveness in bladder required conventional T cells as expected, but not γδ T cells, altogether defining distinct mechanisms for IL-2c and αPD-L1 efficacy. γδ T cells did not improve IL-2c treatment of subcutaneously challenged BC or orthotopic (peritoneal) ovarian cancer, consistent with tissue-specific and/or tumor-specific γδ T cell contributions to IL-2c efficacy. IL-2c significantly altered bladder intratumoral γδ T cell content, activation status, and specific γδ T cell subsets with antitumor or protumor effector functions. Neither IL-2c nor αPD-L1 alone treated lung metastatic MB49 or MBT-2 BC, but their combination improved survival in both models. Combination treatment efficacy in lungs required CD8+ T cells but not γδ T cells. CONCLUSIONS Mechanistic insights into differential IL-2c and αPD-L1 treatment and tissue-dependent effects could help develop rational combination treatment strategies to improve treatment efficacy in distinct cancers. These studies also provide insights into γδ T cell contributions to immunotherapy in bladder and engagement of adaptive immunity by IL-2c plus αPD-L1 to treat refractory lung metastases.
Collapse
Affiliation(s)
- Ryan Michael Reyes
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, San Antonio, Texas, USA
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Yilun Deng
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Deyi Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- National Heart, Blood, and Lung Institute, National Institutes of Health, Bethesda, MD, USA
| | - Niannian Ji
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Neelam Mukherjee
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Karen Wheeler
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Harshita B Gupta
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Alvaro S Padron
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Aravind Kancharla
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Chenghao Zhang
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Myrna Garcia
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Anand V R Kornepati
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, and Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | | | - Robert S Svatek
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, USA
- Department of Urology, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Tyler J Curiel
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas, USA
- Division of Hematology/Oncology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
- Clayton Foundation for Research, Houston, Texas, USA
| |
Collapse
|
21
|
Grivas P, Agarwal N, Pal S, Kalebasty AR, Sridhar SS, Smith J, Devgan G, Sternberg CN, Bellmunt J. Avelumab first-line maintenance in locally advanced or metastatic urothelial carcinoma: Applying clinical trial findings to clinical practice. Cancer Treat Rev 2021; 97:102187. [PMID: 33839438 DOI: 10.1016/j.ctrv.2021.102187] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/21/2022]
Abstract
Although urothelial carcinoma (UC) is considered a chemotherapy-sensitive tumor, progression-free survival and overall survival (OS) are typically short following standard first-line (1L) platinum-containing chemotherapy in patients with locally advanced or metastatic disease. Immune checkpoint inhibitors (ICIs) have antitumor activity in UC and favorable safety profiles compared with chemotherapy; however, trials of 1L ICI monotherapy or chemotherapy + ICI combinations have not yet shown improved OS vs chemotherapy alone. In addition to direct cytotoxicity, chemotherapy has potential immunogenic effects, providing a rationale for assessing ICIs as switch-maintenance therapy. In the JAVELIN Bladder 100 phase 3 trial, avelumab administered as 1L maintenance with best supportive care (BSC) significantly prolonged OS vs BSC alone in patients with locally advanced or metastatic UC that had not progressed with 1L platinum-containing chemotherapy (median OS, 21.4 vs 14.3 months; hazard ratio, 0.69 [95% CI, 0.56-0.86]; P = 0.001). Efficacy benefits were seen across various subgroups, including recipients of 1L cisplatin- or carboplatin-based chemotherapy, patients with PD-L1+ or PD-L1- tumors, and patients with diverse characteristics. Results from JAVELIN Bladder 100 led to the approval of avelumab as 1L maintenance therapy for patients with locally advanced or metastatic UC that has not progressed with platinum-containing chemotherapy. Avelumab 1L maintenance is also included as a standard of care in treatment guidelines for advanced UC with level 1 evidence. This review summarizes the data that supported these developments and discusses practical considerations for administering avelumab maintenance in clinical practice, including patient selection and treatment management.
Collapse
Affiliation(s)
- Petros Grivas
- University of Washington School of Medicine, Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, Seattle, WA, USA.
| | - Neeraj Agarwal
- University of Utah Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Sumanta Pal
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | | | | | - Jodi Smith
- EMD Serono, Inc., Rockland, MA, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | | | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine, Meyer Cancer Center, New York, New York, USA
| | - Joaquim Bellmunt
- Beth Israel Deaconess Medical Center and IMIM-PSMAR Lab, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Wilkins A, Ost P, Sundahl N. Is There a Benefit of Combining Immunotherapy and Radiotherapy in Bladder Cancer? Clin Oncol (R Coll Radiol) 2021; 33:407-414. [PMID: 33726945 DOI: 10.1016/j.clon.2021.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 02/10/2021] [Accepted: 02/25/2021] [Indexed: 01/13/2023]
Abstract
Immune checkpoint inhibitors have transformed the management of patients with metastatic urothelial cancer, by leading to long-term response and prolongation of survival in a subset of patients. Unfortunately, only one in five patients with metastatic urothelial cancer responds to anti-programmed death ligand-1 ([AQ1]anti-PD-1) monotherapy. Preclinical and early clinical evidence indicates that radiotherapy not only acts locally, but also exerts systemic anti-tumour effects by modulating the immune system. It is hypothesised that combining checkpoint inhibitors with radiotherapy might enhance an anti-tumour immune response and increase response rates. So far, a handful of early phase clinical trials have been performed seeking to answer this question in urothelial cancer patients. The current review summarises the available preclinical and clinical evidence on radiotherapy/immunotherapy combinations in locally advanced and metastatic bladder cancer and suggests future avenues worthy of exploration.
Collapse
Affiliation(s)
- A Wilkins
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - P Ost
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium
| | - N Sundahl
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK; Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
23
|
de Jong FC, Rutten VC, Zuiverloon TCM, Theodorescu D. Improving Anti-PD-1/PD-L1 Therapy for Localized Bladder Cancer. Int J Mol Sci 2021; 22:2800. [PMID: 33802033 PMCID: PMC7998260 DOI: 10.3390/ijms22062800] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
In high-risk non-muscle invasive bladder cancer (HR-NMIBC), patient outcome is negatively affected by lack of response to Bacillus-Calmette Guérin (BCG) treatment. Lack of response to cisplatin-based neoadjuvant chemotherapy and cisplatin ineligibility reduces successful treatment outcomes in muscle-invasive bladder cancer (MIBC) patients. The effectiveness of PD-1/PD-L1 immune checkpoint inhibitors (ICI) in metastatic disease has stimulated its evaluation as a treatment option in HR-NMIBC and MIBC patients. However, the observed responses, immune-related adverse events and high costs associated with ICI have provided impetus for the development of methods to improve patient stratification, enhance anti-tumorigenic effects and reduce toxicity. Here, we review the challenges and opportunities offered by PD-1/PD-L1 inhibition in HR-NMIBC and MIBC. We highlight the gaps in the field that need to be addressed to improve patient outcome including biomarkers for response stratification and potentially synergistic combination therapy regimens with PD-1/PD-L1 blockade.
Collapse
Affiliation(s)
- Florus C. de Jong
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Vera C. Rutten
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Tahlita C. M. Zuiverloon
- Department of Urology, Erasmus University Medical Center, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands; (F.C.d.J.); (V.C.R.); (T.C.M.Z.)
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Departments of Surgery (Urology) and Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
24
|
Zhang D, Reyes RM, Osta E, Kari S, Gupta HB, Padron AS, Kornepati AVR, Kancharla A, Sun X, Deng Y, Wu B, Vadlamudi R, Li R, Svatek RS, Curiel TJ. Bladder cancer cell-intrinsic PD-L1 signals promote mTOR and autophagy activation that can be inhibited to improve cytotoxic chemotherapy. Cancer Med 2021; 10:2137-2152. [PMID: 33626233 PMCID: PMC7957205 DOI: 10.1002/cam4.3739] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/16/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
Tumor cell-intrinsic programmed death-ligand 1 (PD-L1) signals mediate immunopathologic effects in breast, colon, and ovarian cancers and in melanomas, but bladder cancer (BC) effects are unreported. We show here that BC cell-intrinsic PD-L1 signals in mouse MB49 and human RT4, UM-UC3, and UM-UC-14 BC cells regulate important pathologic pathways and processes, including effects not reported in other cancers. α-PD-L1 antibodies reduced BC cell proliferation in vitro, demonstrating direct signaling effects. BC cell-intrinsic PD-L1 promoted mammalian target of rapamycin complex 1 (mTORC1) signals in vitro and augmented in vivo immune-independent cell growth and metastatic cancer spread, similar to effects we reported in melanoma and ovarian cancer. BC cell-intrinsic PD-L1 signals also promoted basal and stress-induced autophagy, whereas these signals inhibited autophagy in melanoma and ovarian cancer cells. BC cell-intrinsic PD-L1 also mediated chemotherapy resistance to the commonly used BC chemotherapy agents cis-platinum and gemcitabine and to the mTORC1 inhibitor, rapamycin. Thus, BC cell-intrinsic PD-L1 signals regulate important virulence and treatment resistance pathways that suggest novel, actionable treatment targets meriting additional studies. As a proof-of-concept, we showed that the autophagy inhibitor chloroquine improved cis-platinum treatment efficacy in vivo, with greater efficacy in PD-L1 null versus PD-L1-replete BC.
Collapse
Affiliation(s)
- Deyi Zhang
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
- Present address:
National Institutes of HealthBethesdaMDUSA
| | - Ryan M. Reyes
- Graduate School of Biomedical SciencesUniversity of Texas HealthSan AntonioTXUSA
- Department of Microbiology, Immunology and Molecular GeneticsUniversity Texas HealthSan AntonioTXUSA
- Mays Cancer Center, University of Texas HealthSan AntonioTXUSA
| | - Erica Osta
- Graduate School of Biomedical SciencesUniversity of Texas HealthSan AntonioTXUSA
- Department of Microbiology, Immunology and Molecular GeneticsUniversity Texas HealthSan AntonioTXUSA
| | - Suresh Kari
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
| | | | - Alvaro S. Padron
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
| | - Anand V. R. Kornepati
- Graduate School of Biomedical SciencesUniversity of Texas HealthSan AntonioTXUSA
- Department of Microbiology, Immunology and Molecular GeneticsUniversity Texas HealthSan AntonioTXUSA
| | | | - Xiujie Sun
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
- Present address:
Department of Biochemistry & Molecular MedicineSchool of Medicine & Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Yilun Deng
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
| | - Bogang Wu
- Department of Molecular MedicineUniversity of Texas HealthSan AntonioTXUSA
- Present address:
Department of Biochemistry & Molecular MedicineSchool of Medicine & Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Ratna Vadlamudi
- Mays Cancer Center, University of Texas HealthSan AntonioTXUSA
- Department of Obstetrics and GynecologyUniversity of Texas Health Science CenterSan AntonioTXUSA
| | - Rong Li
- Mays Cancer Center, University of Texas HealthSan AntonioTXUSA
- Department of Molecular MedicineUniversity of Texas HealthSan AntonioTXUSA
- Present address:
Department of Biochemistry & Molecular MedicineSchool of Medicine & Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Robert S. Svatek
- Mays Cancer Center, University of Texas HealthSan AntonioTXUSA
- Department of UrologyUniversity of Texas Health Science CenterSan AntonioTXUSA
| | - Tyler J. Curiel
- Department of MedicineUniversity of Texas HealthSan AntonioTXUSA
- Graduate School of Biomedical SciencesUniversity of Texas HealthSan AntonioTXUSA
- Department of Microbiology, Immunology and Molecular GeneticsUniversity Texas HealthSan AntonioTXUSA
- Mays Cancer Center, University of Texas HealthSan AntonioTXUSA
| |
Collapse
|
25
|
Xing Y, Liu J, Sun S, Ming T, Wang Y, Luo J, Xiao G, Li X, Xie J, Cai X. New electrochemical method for programmed death-ligand 1 detection based on a paper-based microfluidic aptasensor. Bioelectrochemistry 2021; 140:107789. [PMID: 33677221 DOI: 10.1016/j.bioelechem.2021.107789] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/02/2021] [Accepted: 02/13/2021] [Indexed: 12/22/2022]
Abstract
As programmed death-ligand 1 (PD-L1) is considered a referenced therapeutic biomarker, a rapid and low-cost method to detect PD-L1 in body fluids is necessary. In this work, a paper-based microfluidic aptasensor for label-free electrochemical detection of PD-L1 in liquids was fabricated. The aptasensor integrates a reaction cell and a three-electrode system, and a differential pulse voltammetry electrochemical method was adopted. PD-L1 aptamer with a low equilibrium dissociation constant was used as a biorecognition molecule. To bind the aptamer and assist in the electrochemical measurement, nanocomposites were synthesized and used to modify the working electrode, which was composed of an amine-functionalized single-walled carbon nanotube, new methylene blue and gold nanoparticles. The basic performance of the aptasensor was tested in phosphate-buffered saline: the linear range was between 10 pg mL-1 and 2.5 ng mL-1, and the detection limit was 10 pg mL-1 (signal/noise = 3). Moreover, the aptasensor was used for the detection of serum samples and compared with an enzyme linked immunosorbent assay (ELISA). The results showed that the aptasensor provides a new low-cost, portable and highly sensitive detection method for PD-L1, as an alternative to ELISA.
Collapse
Affiliation(s)
- Yu Xing
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Shuai Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Tao Ming
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yang Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guihua Xiao
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinrong Li
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jingyu Xie
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
26
|
Shiga M, Miyazaki J, Tanuma K, Nagumo Y, Yoshino T, Kandori S, Negoro H, Kojima T, Tanaka R, Okiyama N, Fujisawa Y, Watanabe M, Yamasaki S, Kiyohara H, Watanabe M, Sato TA, Tahara H, Nishiyama H, Yano I. The liposome of trehalose dimycolate extracted from M. bovis BCG induces antitumor immunity via the activation of dendritic cells and CD8 + T cells. Cancer Immunol Immunother 2021; 70:2529-2543. [PMID: 33570675 DOI: 10.1007/s00262-021-02870-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/20/2021] [Indexed: 10/22/2022]
Abstract
Intravesical Bovis bacillus Calmette-Guérin (BCG) therapy is the most effective immunotherapy for bladder cancer, but it sometime causes serious side effects because of its inclusion of live bacteria. It is necessary to develop a more active but less toxic immunotherapeutic agent. Trehalose 6,6'-dimycolate (TDM), the most abundant hydrophobic glycolipid of the BCG cell wall, has been reported to show various immunostimulatory activities such as granulomagenesis and adjuvant activity. Here, we developed cationic liposomes incorporating TDM purified from Mycobacterium bovis BCG Connaught, and we investigated the antitumor effect of the cationic liposome TDM (Lip-TDM). Lip-TDM exerted an antitumor effect in bladder cancer, colon cancer, and melanoma-bearing mouse models that was comparable or even superior to that of BCG, with no body weight loss or granuloma formation. The antitumor effect of Lip-TDM disappeared in two types of mice: those with depletion of CD8+ T cells, and those with knockout of macrophage-inducible C-type lectin (Mincle) which recognize TDM. Lip-TDM treatment enhanced the maturation and migration of dendritic cells in the tumor microenvironment in a Mincle-dependent manner. Our results elucidate mechanisms that underlie Lip-TDM treatment and suggest that Lip-TDM has potential as a safe and effective treatment for various cancers.
Collapse
Affiliation(s)
- Masanobu Shiga
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Jun Miyazaki
- Department of Urology, School of Medicine, International University of Health and Welfare, 852 Hatakeda, Narita, Chiba, 286-0124, Japan.
| | - Kozaburo Tanuma
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yoshiyuki Nagumo
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takayuki Yoshino
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shuya Kandori
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hiromitsu Negoro
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takahiro Kojima
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ryota Tanaka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoko Okiyama
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Miyuki Watanabe
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Sho Yamasaki
- Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan.,Laboratory of Molecular Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Osaka, Japan.,Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Japan.,Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chuo-ku, Chiba, Japan
| | | | - Makoto Watanabe
- Life Science Research Center, Technology Research Laboratory, Shimadzu, Kyoto, Japan
| | - Taka-Aki Sato
- Life Science Research Center, Technology Research Laboratory, Shimadzu, Kyoto, Japan
| | - Hideaki Tahara
- Project Division of Cancer Biomolecular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Department of Cancer Drug Discovery and Development, Osaka International Cancer Center, Osaka, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
27
|
Park K, Özgüroğlu M, Vansteenkiste J, Spigel D, Yang JCH, Bajars M, Ruisi M, Manitz J, Barlesi F. Impact of subsequent immune checkpoint inhibitor treatment on overall survival with avelumab vs docetaxel in platinum-treated advanced NSCLC: Post hoc analyses from the phase 3 JAVELIN Lung 200 trial. Lung Cancer 2021; 154:92-98. [PMID: 33636453 DOI: 10.1016/j.lungcan.2021.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 01/24/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The JAVELIN Lung 200 phase 3 trial did not meet its primary endpoint of improving overall survival (OS) with avelumab vs docetaxel in patients with platinum-treated PD-L1+ NSCLC. We report post hoc analyses assessing the effects of subsequent immune checkpoint inhibitor (ICI) treatment on OS. MATERIAL AND METHODS Patients with stage IIIB/IV NSCLC progressed following platinum-doublet therapy were randomized to receive avelumab or docetaxel. OS was analyzed in the PD-L1+ population (≥1% of tumor cells) and full analysis set (PD-L1+ or PD-L1-). Effects of subsequent ICI (after permanent discontinuation of study treatment) on OS were analyzed using a preplanned naive sensitivity analysis and post hoc inverse probability of censoring weighting (IPCW) analysis. Subgroups with or without subsequent ICI treatment were analyzed using descriptive statistics. RESULTS In the avelumab and docetaxel arms, a subsequent ICI was received by 16/396 (4.0 %) and 104/396 (26.3 %) after a median of 10.5 months (range, 3.9-20.4) and 5.7 months (range, 0.1-24.4), respectively. Some subgroups showed trends for higher subsequent ICI treatment, including patients with non-squamous NSCLC (avelumab arm, 4.3 % vs docetaxel arm, 32.1 %) or with a baseline ECOG performance status of 0 (6.3 % vs 31.3 %); those enrolled in the early recruitment wave (11.6 % vs 54.3 %), or enrolled in the US/Western Europe (2.8 % vs 45.5 %) or Asia (11.0 % vs 35.4 %); and non-white patients (10.1 % vs 35.0 %). The hazard ratio for OS with avelumab vs docetaxel was lower in the IPCW analysis than in the naive sensitivity analysis (PD-L1+ population: 0.80 [95 % CI, 0.62-1.04] vs 0.86 [95 % CI, 0.68-1.09], respectively). CONCLUSION In the JAVELIN Lung 200 trial, avelumab showed clinical activity as second-line treatment for patients with advanced NSCLC. Post hoc analyses suggest that the primary OS analysis may have been confounded by subsequent ICI use in the docetaxel arm. ClinicalTrials.gov identifier: NCT02395172.
Collapse
Affiliation(s)
- Keunchil Park
- Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Mustafa Özgüroğlu
- Department of Internal Medicine, Division of Medical Oncology, Cerrahpaşa Medical Faculty, Istanbul University Cerrahpaşa, Istanbul, Turkey.
| | - Johan Vansteenkiste
- Department of Respiratory Oncology, University Hospital KU Leuven, Leuven, Belgium.
| | - David Spigel
- Sarah Cannon Research Institute, Nashville, TN, USA.
| | - James C-H Yang
- Department of Medical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
| | - Marcis Bajars
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA; an affiliate of Merck KGaA, Darmstadt, Germany.
| | - Mary Ruisi
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA; an affiliate of Merck KGaA, Darmstadt, Germany.
| | - Juliane Manitz
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA; an affiliate of Merck KGaA, Darmstadt, Germany.
| | - Fabrice Barlesi
- Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France; Gustave Roussy Cancer Campus, Villejuif, France.
| |
Collapse
|
28
|
Novel Adjuvant Therapies for Upper Tract Urothelial Carcinoma After Endoscopic Management. Bladder Cancer 2021. [DOI: 10.1007/978-3-030-70646-3_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
29
|
Rousseau M, O’Brien CJ, Antequera E, Zdimerova H, Cansever D, Canton T, Zychlinsky Scharff A, Ingersoll MA. Identification of Sex Differences in Tumor-Specific T Cell Infiltration in Bladder Tumor-Bearing Mice Treated with BCG Immunotherapy. Bladder Cancer 2020. [DOI: 10.3233/blc-200384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND: Bladder cancer is the fourth most common cancer for men. However, women are often diagnosed with later stage disease and have poorer outcomes. Whether immune-based sex differences contribute to this discrepancy is unclear. In addition, models to investigate tumor-specific immunity in bladder cancer, in the context of tumor development or response to therapy, are lacking. OBJECTIVE: To address this specific unmet need, we incorporated a commonly used model antigen, ovalbumin, into two well-established models of bladder cancer; the orthotopic MB49 cell line model and the carcinogenic BBN bladder cancer model. METHOD: We tested the utility of these models to investigate tumor-specific immunity in the context of immunotherapy in both sexes. RESULTS: We found that BCG vaccination, prior to weekly BCG instillation does not impart an immune-specific benefit to tumor-bearing mice in the context of multiple BCG instillations. Furthermore, tumors developed in the testes in male mice, precluding the use of the MB49 model to directly investigate sex-based immune differences. In the BBN model, we observed that more tumor antigen-specific CD8+ T cells infiltrated male bladders compared to female bladders in the context of BCG immunotherapy whereas regulatory T cells had higher levels of the exhaustion marker PD-1 in female mice. CONCLUSIONS: We propose our modified BBN model will contribute to our understanding of how tumor-specific immunity arises in bladder cancer. Additionally, the BBN bladder cancer model may help to uncover sex differences in tumor-specific immunity, which would provide valuable information for the development of new treatments or combination therapies for bladder cancer in women and men.
Collapse
Affiliation(s)
- Matthieu Rousseau
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Conan J.O. O’Brien
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Eduardo Antequera
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Hana Zdimerova
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Dilay Cansever
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | - Tracy Canton
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| | | | - Molly A. Ingersoll
- Department of Immunology, Institut Pasteur, Paris, France
- INSERM U1223, Paris, France
| |
Collapse
|
30
|
Rouanne M, Bajorin DF, Hannan R, Galsky MD, Williams SB, Necchi A, Sharma P, Powles T. Rationale and Outcomes for Neoadjuvant Immunotherapy in Urothelial Carcinoma of the Bladder. Eur Urol Oncol 2020; 3:728-738. [PMID: 33177001 DOI: 10.1016/j.euo.2020.06.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/14/2020] [Accepted: 06/24/2020] [Indexed: 12/17/2022]
Abstract
CONTEXT Immune therapy has emerged as a powerful treatment of metastatic urothelial carcinoma. Over 20 ongoing studies are exploring this strategy in the neoadjuvant setting in patients with localized muscle-invasive bladder cancer. OBJECTIVE To summarize the rationale and the clinical outcomes regarding the use of immune checkpoint blockade in the neoadjuvant setting before radical cystectomy. EVIDENCE ACQUISITION A systematic review of the literature in the MEDLINE database was performed. The central search strategy used the terms bladder cancer, urothelial carcinoma, mice, human, immunotherapy, neoadjuvant therapy, atezolizumab, pembrolizumab, durvalumab, nivolumab, avelumab, ipilimumab, and tremelimumab. The search was limited to publications between January 2008 and February 2020. Publicly available relevant abstracts from recent meetings were also included. EVIDENCE SYNTHESIS Phase II trials investigating neoadjuvant immune checkpoint blockade as a single agent before radical cystectomy reported a rate of pathological complete response (CR), ranging from 31% with an anti-PD-L1 monoclonal antibody (mAb) atezolizumab (n = 27/88) to 37% with anti-PD-1 mAb pembrolizumab (n = 42/114). Overall, 92% (n = 87/95) and 98% (n = 112/114) of the patients underwent radical cystectomy. Neoadjuvant immune checkpoint blockade did not delay planned surgery. Checkpoint inhibitor monotherapy was well tolerated, with no unexpected toxicity in the presurgical setting. Early phase I/II trials investigating neoadjuvant combination chemotherapy strategies with immune checkpoint blockers reported enhanced antitumor efficacy, with a pathological CR ranging from 33% to 50%. CONCLUSIONS Although limited clinical data are available on long-term survival, neoadjuvant immune checkpoint blockade demonstrated effective antitumor efficacy for localized muscle-invasive bladder cancer. Phase III trials are currently investigating this strategy in the presurgical setting. PATIENT SUMMARY Immunotherapy prior to surgery has been evaluated for patients with muscle-invasive bladder cancer. Although long-term survival benefit is unknown, such treatment strategy revealed a promising antitumor response rate for patients who underwent radical cystectomy. Ongoing prospective clinical trials will define the potential advantage of this approach over current cisplatin-based chemotherapeutic regimens alone or in combination.
Collapse
Affiliation(s)
- Mathieu Rouanne
- Department of Urology, Hôpital Foch, UVSQ-Université Paris-Saclay, Suresnes, France; INSERM U1015, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Dean F Bajorin
- Department of Medicine, Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raquibul Hannan
- Department of Radiation Oncology, Harold C. Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew D Galsky
- Division of Hematology and Oncology, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stephen B Williams
- Division of Urology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Andrea Necchi
- Department of Medical Oncology, Fondazione IRCCS Instituto Nazionale dei Tumori, Milan, Italy
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Powles
- Experimental Cancer Medicine Centre Barts Cancer Institute, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| |
Collapse
|
31
|
Assessment of prognostic implication of a panel of oncogenes in bladder cancer and identification of a 3-gene signature associated with recurrence and progression risk in non-muscle-invasive bladder cancer. Sci Rep 2020; 10:16641. [PMID: 33024200 PMCID: PMC7538919 DOI: 10.1038/s41598-020-73642-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
This study evaluated the prognostic value of a panel of 29 oncogenes derived from the analysis of The Cancer Genome Atlas (TCGA data) or from the recent literature on bladder tumors on a well-characterized series of muscle-invasive bladder cancer (MIBC) and non-MIBC (NMIBC) samples and tried to identify molecular prognostic markers. Mutations of HRAS, FGFR3, PIK3CA and TERT were found in 2.9%, 27.2%, 14.9% and 76.7% of tumor samples, respectively. Concerning NMIBC, on multivariate analysis, RXRA and FGFR3 levels were associated with recurrence-free survival (RFS) (p = 0.0022 and p = 0.0069) and RXRA level was associated with progression to muscle-invasive disease (p = 0.0068). We identified a 3-gene molecular signature associated with NMIBC prognosis. FGFR3 overexpression was associated with reduced response to Bacillus Calmette–Guerin treatment (p = 0.037). As regards MIBC, on multivariate analysis, ERCC2 overexpression was associated with RFS (p = 0.0011) and E2F3 and EGFR overexpression were associated with overall survival (p = 0.014 and p = 0.035). RT-PCR findings were confirmed by IHC for FGFR3. Genomic alterations in MIBC revealed in TCGA data also concern NMIBC and seem to be associated with prognosis in terms of recurrence and progression. Correcting these alterations by targeted therapies seems a promising pharmacological approach.
Collapse
|
32
|
Verschraegen CF, Jerusalem G, McClay EF, Iannotti N, Redfern CH, Bennouna J, Chen FL, Kelly K, Mehnert J, Morris JC, Taylor M, Spigel D, Wang D, Grote HJ, Zhou D, Munshi N, Bajars M, Gulley JL. Efficacy and safety of first-line avelumab in patients with advanced non-small cell lung cancer: results from a phase Ib cohort of the JAVELIN Solid Tumor study. J Immunother Cancer 2020; 8:jitc-2020-001064. [PMID: 32907924 PMCID: PMC7481079 DOI: 10.1136/jitc-2020-001064] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2020] [Indexed: 02/06/2023] Open
Abstract
Introduction Avelumab, an antiprogrammed death ligand-1 antibody, is approved as a monotherapy for treatment of metastatic Merkel cell carcinoma and advanced urothelial carcinoma, and in combination with axitinib for advanced renal cell carcinoma. We report the efficacy and safety of first-line avelumab in advanced non-small cell lung cancer (NSCLC). Methods In a phase I expansion cohort of the JAVELIN Solid Tumor trial, patients with treatment-naive, metastatic, or recurrent NSCLC received 10 mg/kg avelumab intravenously every 2 weeks. Endpoints included best overall response, duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. Results Overall, 156 patients were enrolled and treated. Median duration of follow-up was 18.6 months (range, 15 to 23 months). The objective response rate was 19.9% (95% CI, 13.9 to 27.0), including complete response in 3 (1.9%) and partial response in 28 (17.9%). Median DOR was 12.0 months (95% CI, 6.9 to not estimable). Median PFS was 4.0 months (95% CI, 2.7 to 5.4) and the 6-month PFS rate was 38.5% (95% CI, 30.7 to 46.3). Median OS was 14.1 months (95% CI, 11.3 to 16.9) and the 12-month OS rate was 56.6% (95% CI, 48.2 to 64.1). Treatment-related adverse events (TRAEs) occurred in 107 patients (68.6%), including grade ≥3 TRAEs in 19 (12.2%). Immune-related adverse events and infusion-related reactions occurred in 31 (19.9%) and 40 patients (25.6%), respectively. No treatment-related deaths occurred. Conclusion Avelumab showed antitumor activity with a tolerable safety profile as a first-line treatment in patients with advanced NSCLC. These data support further investigation of avelumab in the phase III JAVELIN Lung 100 study. Trial registration details ClinicalTrials.gov NCT01772004; registered January 21, 2013.
Collapse
Affiliation(s)
- Claire F Verschraegen
- Division of Medical Oncology, The Ohio State University James Cancer Hospital, Columbus, Ohio, USA
| | - Guy Jerusalem
- Department of Medical Oncology, CHU Sart Tilman Liege and Liege University, Liege, Belgium
| | - Edward F McClay
- Institute for Melanoma Research & Education, California Cancer Associates for Research & Excellence, Encinitas, California, USA
| | - Nicholas Iannotti
- Hematology Oncology Associates of the Treasure Coast, Port St. Lucie, Florida, USA
| | | | - Jaafar Bennouna
- Department of Pneumology, Thoracic Oncology, University Hospital Centre Nantes, Nantes, France
| | - Franklin L Chen
- Novant Health Oncology Specialists, Winston-Salem, North Carolina, USA
| | - Karen Kelly
- Internal Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Janice Mehnert
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
| | - John C Morris
- University of Cincinnati Cancer Institute, Cincinnati, Ohio, USA
| | | | - David Spigel
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| | - Ding Wang
- Henry Ford Medical Center, Detroit, Michigan, USA
| | | | - Dongli Zhou
- Merck Serono Pharmaceutical R&D Co, Beijing, China; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Neru Munshi
- EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA; a business of Merck KGaA, Darmstadt, Germany
| | - Marcis Bajars
- EMD Serono Research & Development Institute, Inc, Billerica, Massachusetts, USA; a business of Merck KGaA, Darmstadt, Germany
| | - James L Gulley
- Genitourinary Malignancies Branch and Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
33
|
Guo H, Li F, Qiu H, Xu W, Li P, Hou Y, Ding J, Chen X. Synergistically Enhanced Mucoadhesive and Penetrable Polypeptide Nanogel for Efficient Drug Delivery to Orthotopic Bladder Cancer. RESEARCH 2020; 2020:8970135. [PMID: 32832909 PMCID: PMC7420878 DOI: 10.34133/2020/8970135] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 03/26/2020] [Indexed: 11/06/2022]
Abstract
Intravesical chemotherapy has been recommended after the gold standard of transurethral resection of the bladder tumor to prevent bladder cancer (BC) from local recurrence in the clinic. However, due to rapid urine excretion and barrier protection of the bladder wall, the clinical performances of chemotherapeutic drugs are severely compromised. In the present work, a smart positively charged disulfide-crosslinked nanogel of oligoarginine-poly(ethylene glycol)–poly(L-phenylalanine-co-L-cystine) (R9-PEG–P(LP-co-LC)) was prepared to prolong the retention period and enhance the penetration capability of chemotherapeutic agent toward the bladder wall. PEG significantly improved the aqueous dispersibility of the 10-hydroxycamptothecin (HCPT)-loaded R9-PEG–P(LP-co-LC) (i.e., R9NG/HCPT) and enhanced the mucoadhesive capability by the nonspecific interaction between PEG chain and the bladder mucosa accompanied with the electrostatic interaction between the cationic R9 and negatively charged bladder mucosa. Besides, R9, as a cell-penetrating peptide, efficiently penetrated through the cell membrane and delivered carried cargo. The disulfide bond endowed the selective release behavior of HCPT triggered by the intracellular reductive microenvironment. As an advanced chemotherapeutic nanoformulation, the smart R9NG/HCPT demonstrated superior cytotoxicity against human BC 5637 cells in vitro and remarkably enhanced tumor suppression activity toward orthotopic BC models of mouse and rat in vivo, indicating its great potential in the clinical intravesical BC chemotherapy.
Collapse
Affiliation(s)
- Hui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China.,Department of Urinary Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Faping Li
- Department of Urinary Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Heping Qiu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China.,Department of Urinary Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Weiguo Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Pengqiang Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Yuchuan Hou
- Department of Urinary Surgery, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| |
Collapse
|
34
|
Abstract
Local and systemic immunotherapies are used for the management of bladder cancer in daily practice. BCG has been administered for almost 40 years in patients with non-muscle invasive bladder cancer (NMIBC). Despite its efficacy, disease progression is observed in nearly 30% of patients. Given the antitumor activity of immune checkpoint inhibitors in metastatic setting, these therapies are currently investigated in NMIBC. Pembrolizumab is now approved by the Food and Drug Administration (FDA) for the treatment of patients with BCG-unresponsive, high-risk, NMIBC with carcinoma in situ with or without papillary tumors who are ineligible for or have not elected to undergo cystectomy. Several phase 3 trials are ongoing to investigate the efficacy of PD(L)1 inhibitors combined with BCG such as ALBAN study in France.
Collapse
|
35
|
Yang Z, Wang H, Zhang N, Xing T, Zhang W, Wang G, Li C, Yu C. Chaetocin Abrogates the Self-Renewal of Bladder Cancer Stem Cells via the Suppression of the KMT1A-GATA3-STAT3 Circuit. Front Cell Dev Biol 2020; 8:424. [PMID: 32626701 PMCID: PMC7311639 DOI: 10.3389/fcell.2020.00424] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022] Open
Abstract
Bladder cancer stem cells (BCSCs) have the abilities of self-renewal, differentiation, and metastasis; confer drug resistance; and exhibit high tumorigenicity. We previously identified that the KMT1A–GATA3–STAT3 axis drives the self-renewal of BCSCs. However, the therapeutic effect of targeting KMT1A in BCSCs remains unknown. In this study, we confirmed that the expression of KMT1A was remarkably higher in BCSCs (3–5-fold) than those in bladder cancer non-stem cells or normal bladder epithelial cells. Among the six KMT1A inhibitors, chaetocin significantly suppressed the cell propagation (inhibition ratio: 65%–88%, IC50 = 24.4–32.5 nM), induced apoptosis (2–5-fold), and caused G1 phase cell cycle arrest (68.9 vs 55.5%) of bladder cancer (BC) cells, without influencing normal bladder epithelial cells. More importantly, chaetocin abrogated the self-renewal of BCSCs (inhibition ratio: 80.1%) via the suppression of the KMT1A–GATA3–STAT3 circuit and other stemness-related pathways. Finally, intravesical instillation of chaetocin remarkably inhibited the growth of xenograft tumors (inhibition ratio: 71–82%) and prolonged the survival of tumor-bearing mice (70 vs 53 days). In sum, chaetocin abrogated the stemness maintenance and tumor growth of BCSCs via the suppression of the KMT1A–GATA3–STAT3 circuit. Chaetocin is an effective inhibitor targeting KMT1A in BCSCs and could be a promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Zhao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Haifeng Wang
- Department of Urology, Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Nan Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Tianying Xing
- Department of Urology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Urology, Affiliated Hospital of Hebei University, Baoding, China
| | - Guoqing Wang
- Department of Pathogenobiology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Chong Li
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
36
|
Vitale LA, He LZ, Thomas LJ, Wasiuk A, O'Neill T, Widger J, Crocker A, Mills-Chen L, Forsberg E, Weidlick J, Patterson C, Hammond RA, Boyer J, Sisson C, Alvarado D, Goldstein J, Marsh HC, Keler T. Development of CDX-527: a bispecific antibody combining PD-1 blockade and CD27 costimulation for cancer immunotherapy. Cancer Immunol Immunother 2020; 69:2125-2137. [PMID: 32451681 PMCID: PMC7511290 DOI: 10.1007/s00262-020-02610-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022]
Abstract
CD27 is a costimulatory molecule that provides a complementary target to the PD-1/PD-L1 checkpoint axis on T cells. Combining a CD27 agonist antibody with PD-1/PD-L1 blockade has shown synergistic antitumor activity in preclinical models, which led to clinical studies of the combination in cancer patients. We theorized that coupling CD27 costimulation with PD-1/PD-L1 blockade in a bispecific antibody (BsAb) may provide greater immune activating properties than combining the individual mAbs due to enhanced CD27 activation by cross-linking through PD-L1 and Fc receptors. To test this approach, we developed CDX-527, a tetravalent PD-L1xCD27 IgG1-scFv BsAb. CDX-527 potently inhibits PD-1 signaling and induces CD27-mediated T cell costimulation through PD-L1 cross-linking. In mixed lymphocyte reaction assays, CDX-527 is more potent than the combination of the parental antibodies, suggesting that cross-linking through both Fc receptors and PD-L1 results in enhanced CD27 agonist activity. CDX-527 was shown to mediate effector function against tumor cells overexpressing either CD27 or PD-L1. In human CD27 transgenic mice, we observed that antigen-specific T cell responses to a vaccine are greatly enhanced with a surrogate PD-L1xCD27 BsAb. Furthermore, the BsAb exhibits greater antitumor activity than the combination of the parental antibodies in a syngeneic lymphoma model. A pilot study of CDX-527 in cynomolgus macaques confirmed a mAb-like pharmacokinetic profile without noted toxicities. These studies demonstrate that CDX-527 effectively combines PD-1 blockade and CD27 costimulation into one molecule that is more potent than combination of the parental antibodies providing the rationale to advance this BsAb toward clinical studies in cancer patients.
Collapse
Affiliation(s)
- Laura A Vitale
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Li-Zhen He
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Lawrence J Thomas
- Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - Anna Wasiuk
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Thomas O'Neill
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Jenifer Widger
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Andrea Crocker
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Laura Mills-Chen
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Eric Forsberg
- Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - Jeffrey Weidlick
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Colleen Patterson
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Russell A Hammond
- Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - James Boyer
- Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - Crystal Sisson
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Diego Alvarado
- Celldex Therapeutics, Inc., 300 George Street, Suite 530, New Haven, CT, 06511, USA
| | - Joel Goldstein
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA
| | - Henry C Marsh
- Celldex Therapeutics, Inc., 151 Martine Street, Fall River, MA, 02723, USA
| | - Tibor Keler
- Celldex Therapeutics, Inc., 53 Frontage Road, Suite 220, Hampton, NJ, 08827, USA.
| |
Collapse
|
37
|
Li M, Ehlerding EB, Jiang D, Barnhart TE, Chen W, Cao T, Engle JW, Cai W. In vivo characterization of PD-L1 expression in breast cancer by immuno-PET with 89Zr-labeled avelumab. Am J Transl Res 2020; 12:1862-1872. [PMID: 32509182 PMCID: PMC7270013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/20/2020] [Indexed: 06/11/2023]
Abstract
Programmed death protein 1 and programmed death-ligand 1 (PD-1/PD-L1) have been widely studied as one of the most critical immune check-point pairs in the cancer microenvironment. In breast cancer (BrCa), the expression of PD-L1 is regarded as a determinant biomarker for patient stratification and prediction of inhibition response. Quantitative positron emission tomography (PET) imaging of PD-L1 expression in tumors using a therapeutic antibody in the clinic seems to be a promising approach that can complement conventional histopathological methods and overcome several issues, such as the tumor heterogeneities, sampling representativeness and clear differentiation of positive and negative results. In this study, we synthesized and evaluated 89Zr-labeled avelumab (Ave) for the in vivo characterization of PD-L1 expression in BrCa. Confocal imaging of BrCa cells and flow cytometry were employed to evaluate PD-L1 expression in MDA-MB-231 cells. The intact human monoclonal antibody targeting PD-L1, i.e., Ave, was conjugated to p-SCN-Deferoxamine (Df) and labeled with 89Zr. After intravenous injection of 89Zr-Df-avelumab (89Zr-Df-Ave), PET imaging of MDA-MB-231 tumor-bearing mice, with or without blocking, was performed. High PD-L1 expression of MDA-MB-231 cells was confirmed by in vitro immuno-fluorescent staining and flow cytometry. PET imaging indicated the peak uptake of 89Zr-Df-Ave in the tumor (6.4±1.0 %ID/g), spleen (10.2±0.7 %ID/g) and lymph nodes (6.9±1.0 %ID/g) at 48 h after injection (n=4). Blocking study using unlabeled Ave could reduce the tracer uptake in these tissues (5.2±1.0 %ID/g in the tumor, 4.9±0.5 %ID/g in the spleen and 5.8±1.1 %ID/g in lymph nodes at 48 h, n=4), which demonstrated the specificity of 89Zr-Df-Ave. Biodistribution study and immuno-fluorescent staining were consistent with the quantitative data from PET imaging. Herein, we offer the evidence supporting the value of immuno-PET imaging using 89Zr-Df-Ave for non-invasive characterization of PD-L1 expression in BrCa and the applicability of this tracer in BrCa for treatment evaluation after immunotherapy.
Collapse
Affiliation(s)
- Miao Li
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University277 West Yanta Road, Xi’an 710061, Shaanxi, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Emily B Ehlerding
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Dawei Jiang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Todd E Barnhart
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Weiyu Chen
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Tianye Cao
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison1111 Highland Avenue, Madison 53705, Wisconsin, United States
| |
Collapse
|
38
|
Waked R, Choucair J, Chehata N, Haddad E, Saliba G. Intravesical Bacillus Calmette-Guérin (BCG) treatment's severe complications: A single institution review of incidence, presentation and treatment outcome. J Clin Tuberc Other Mycobact Dis 2020; 19:100149. [PMID: 32099909 PMCID: PMC7016447 DOI: 10.1016/j.jctube.2020.100149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Intravesical Bacillus Calmette-Guérin (BCG) treatment for superficial bladder cancer is interrupted in approximatively 8% of cases as a result of complications. The objective is to report the severe related complications of Bacillus Calmette-Guérin (BCG) following an intravesical instillation for bladder tumor encountered at our institution for the past 5 years. METHODS Medical records of a tertiary teaching hospital, located in Beirut, Lebanon, were retrospectively analyzed from June 2014 to June 2019 searching for severe related complications of BCG. A comprehensive review of articles on this subject was conducted. RESULTS The incidence of severe systemic adverse events related to BCG instillation was 1.5% (5 out of 332 patients). A total of five patients were found to have a severe BCG related complication, with fever, chills, and irritative urinary signs being the most frequent symptoms. All patients received antituberculosis therapy (Isoniazid, Rifampin and Ethambutol). Two were put on add-on corticosteroids. Three patients had a computed tomography scan image in favor of an infection. Two patients had a favorable outcome, three patients died. CONCLUSION BCG severe adverse events were mostly seen in patients with a traumatic instillation. Treatment used at our institution was similar to most cases reported in the literature. A standardized diagnostic and treatment approach should be implemented to help physicians tackle these life-threatening complications.
Collapse
Affiliation(s)
- R. Waked
- Department of Infectious Diseases, Faculty of Medicine, Saint Joseph University, Damascus street, PO BOX 11-5076, Riad El Solh, Beirut 1107 2180, Lebanon
| | - J. Choucair
- Coordinator of the Infectious Diseases department, Saint Joseph University, Beirut, Lebanon
| | - N. Chehata
- Department of Infectious Diseases, Faculty of Medicine, Saint Joseph University, Damascus street, PO BOX 11-5076, Riad El Solh, Beirut 1107 2180, Lebanon
| | - E. Haddad
- Department of Infectious Diseases, Faculty of Medicine, Saint Joseph University, Damascus street, PO BOX 11-5076, Riad El Solh, Beirut 1107 2180, Lebanon
| | - G. Saliba
- Department of Infectious Diseases, Faculty of Medicine, Saint Joseph University, Damascus street, PO BOX 11-5076, Riad El Solh, Beirut 1107 2180, Lebanon
| |
Collapse
|
39
|
Grote HJ, Feng Z, Schlichting M, Helwig C, Ruisi M, Jin H, Scheuenpflug J, Gann CN, Su Z, Reck M, Vokes EE, Kerr KM. Programmed Death-Ligand 1 Immunohistochemistry Assay Comparison Studies in NSCLC: Characterization of the 73-10 Assay. J Thorac Oncol 2020; 15:1306-1316. [PMID: 32353599 DOI: 10.1016/j.jtho.2020.04.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Several programmed death-ligand 1 (PD-L1) immunohistochemistry (IHC) assays have been developed independently within clinical programs for therapeutic anti-programmed cell death protein 1 (anti-PD-1) or PD-L1 antibodies, necessitating assessment of assay comparability. We characterized the Dako PD-L1 IHC 73-10 assay used in clinical trials of avelumab (anti-PD-L1) or bintrafusp alfa (M7824; bifunctional immunotherapy) and compared it with the Dako PD-L1 IHC 22C3 pharmDx assay, an approved companion diagnostic for pembrolizumab monotherapy in patients with advanced NSCLC. METHODS Formalin-fixed, paraffin-embedded NSCLC tumor samples from a commercial source and from the JAVELIN Solid Tumor phase 1 trial of avelumab (NCT01772004) were stained using the 73-10 and 22C3 IHC assays with a standard protocol. RESULTS Both assays displayed expected PD-L1 staining patterns. In 148 commercial NSCLC samples, the 73-10 assay stained greater than or equal to 1%, greater than or equal to 50%, and greater than or equal to 80% of tumor cells as PD-L1+ in 64.2%, 36.5%, and 23.6% of the samples, respectively, whereas the 22C3 assay stained 20.3% of the samples as greater than or equal to 50% PD-L1+. In 83 NSCLC clinical trial samples, the 73-10 assay stained 79.5% and 31.3% of the samples as greater than or equal to 1% and greater than or equal to 80% PD-L1+, respectively, whereas the 22C3 assay stained 59.0% and 21.7% as greater than or equal to 1% and greater than or equal to 50% PD-L1+, respectively. Efficacy of avelumab was similar in the subgroups classified with the 73-10 and 22C3 assays using greater than or equal to 80% and greater than or equal to 50% PD-L1+ cutoffs, with objective response rates of 26.9% and 33.3%, respectively. CONCLUSIONS The 73-10 assay demonstrated high sensitivity for PD-L1 staining, and staining was comparable between the greater than or equal to 80% cutoff of the 73-10 assay and greater than or equal to 50% cutoff of the 22C3 assay.
Collapse
Affiliation(s)
| | - Zheng Feng
- EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts; a business of Merck KGaA, Darmstadt, Germany
| | | | | | - Mary Ruisi
- EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts; a business of Merck KGaA, Darmstadt, Germany
| | | | | | | | - Zhen Su
- EMD Serono Research & Development Institute, Inc., Rockland, Massachusetts; a business of Merck KGaA, Darmstadt, Germany
| | - Martin Reck
- Lung Clinic, Airway Research Center North, German Center for Lung Research (Deutsches Zentrum für Lungenforschung), Grosshansdorf, Germany
| | - Everett E Vokes
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Keith M Kerr
- Department of Pathology, Aberdeen University Medical School and Aberdeen Royal Infirmary, Aberdeen, United Kingdom.
| |
Collapse
|
40
|
Larsen ES, Joensen UN, Poulsen AM, Goletti D, Johansen IS. Bacillus Calmette-Guérin immunotherapy for bladder cancer: a review of immunological aspects, clinical effects and BCG infections. APMIS 2020; 128:92-103. [PMID: 31755155 DOI: 10.1111/apm.13011] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/10/2019] [Indexed: 12/29/2022]
Abstract
Bacillus Calmette-Guérin (BCG) immunotherapy for bladder cancer has been used since 1976 when the first evidence of its ability to lower recurrence and progression rates was published. Today, BCG immunotherapy is the choice of care for high-grade non-muscle invasive bladder cancer (NMIBC) after transurethral resection. This article presents indications and procedure of BCG instillations, and outlines the effects on recurrence and progression of NMIBC. The BCG-induced immunity in NMIBC is not yet fully understood. Animal studies point towards BCG inducing specific tumour immunity. We describe the current knowledge of how this immunity is induced, from internalization of BCG bacilli in urothelial cells, to cytokine- and chemokine-mediated recruitment of neutrophils, monocytes, macrophages, T cells, B cells and natural killer cells. In addition, we describe the process of trained immunity, the non-specific protective effects of BCG. Recent studies also indicate that dysbiosis of the urinary microbiome may cause lower urinary tract dysfunction. Side effects of BCG bladder instillations range from common, mild and transient symptoms, such as dysuria and flu-like symptoms, to more severe and rarely occurring life-threatening complications. We review the literature and give an overview of reported incidences and management of BCG infections after intravesical instillation.
Collapse
Affiliation(s)
| | - Ulla Nordström Joensen
- Department of Urology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Alicia Martin Poulsen
- Department of Urology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS L. Spallanzani, Rome, Italy
| | - Isik Somuncu Johansen
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Mycobacterial Centre for Research Southern Denmark - MyCRESD, Odense, Denmark
| |
Collapse
|
41
|
Abstract
After Bacillus Calmette-Guerin (BCG) failure, there is likely a 6- to 24-month window whereby salvage intravesical therapy might allow for preservation of the bladder without disease worsening. Combination intravesical, salvage therapy for nonmuscle invasive bladder cancer represents a promising avenue for treatment in patients unfit or unwilling to undergo cystectomy. BCG with concomitant immune stimulating agents or immune checkpoint inhibitors, combination chemotherapy regimens, such as gemcitabine and docetaxol, and novel agents currently in clinical trials provide hope for a bladder-sparing alternative for patients after BCG failure.
Collapse
Affiliation(s)
- Nathan A Brooks
- Department of Urology, The University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, USA
| | - Michael A O'Donnell
- Department of Urology, The University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, USA.
| |
Collapse
|
42
|
Novakovic AM, Wilkins JJ, Dai H, Wade JR, Neuteboom B, Brar S, Bello CL, Girard P, Khandelwal A. Changing Body Weight-Based Dosing to a Flat Dose for Avelumab in Metastatic Merkel Cell and Advanced Urothelial Carcinoma. Clin Pharmacol Ther 2019; 107:588-596. [PMID: 31553054 PMCID: PMC7027979 DOI: 10.1002/cpt.1645] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/17/2019] [Indexed: 01/04/2023]
Abstract
Avelumab, an anti–programmed death‐ligand 1 monoclonal antibody approved for the treatment of metastatic Merkel cell carcinoma and platinum‐treated urothelial carcinoma, was initially approved with a 10 mg/kg weight‐based dose. We report pharmacokinetic (PK)/pharmacodynamic analyses for avelumab comparing weight‐based dosing and a flat 800 mg dose, developed using data from 1,827 patients enrolled in 3 clinical trials (NCT01772004, NCT01943461, and NCT02155647). PK metrics were simulated for weight‐based and flat‐dosing regimens and summarized by quartiles of weight. Derived exposure metrics were used in simulations of exposure‐safety (various tumors) and exposure‐efficacy (objective responses; Merkel cell or urothelial carcinoma). Flat dosing was predicted to provide similar exposure to weight‐based dosing, with slightly lower variability. Exposure‐safety and exposure‐efficacy simulations suggested similar benefit:risk profiles for the two dosing regimens. These pharmacometric analyses provided the basis for the US Food and Drug Administration approval of a flat dose of avelumab 800 mg every 2 weeks in approved indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pascal Girard
- Merck Institute of Pharmacometrics, Merck Serono SA, Lausanne, Switzerland
| | | |
Collapse
|
43
|
Rompré-Brodeur A, Shinde-Jadhav S, Ayoub M, Piccirillo CA, Seuntjens J, Brimo F, Mansure JJ, Kassouf W. PD-1/PD-L1 Immune Checkpoint Inhibition with Radiation in Bladder Cancer: In Situ and Abscopal Effects. Mol Cancer Ther 2019; 19:211-220. [PMID: 31534011 DOI: 10.1158/1535-7163.mct-18-0986] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 04/02/2019] [Accepted: 09/12/2019] [Indexed: 11/16/2022]
Abstract
The combination of radiation with immune checkpoint inhibitors was reported in some cancers to have synergic effects both locally and distally. Our aim was to assess this combined therapy on both radiated and nonradiated bladder tumors and to characterize the immune landscape within the tumor microenvironment. Murine bladder cancer cells (MB49) were injected subcutaneously in both flanks of C57BL/6 mice. Mice were randomly assigned to the following treatments: placebo, anti-PD-L1 (four intraperitoneal injections over 2 weeks), radiation to right flank (10 Gy in two fractions), or radiation+anti-PD-L1. Tumor digestion, flow cytometry, and qPCR were performed. Log-rank analysis was used for statistical significance. Radiation+anti-PD-L1 group demonstrated statistically significant slower tumor growth rate both in the radiated and nonirradiated tumors (P < 0.001). Survival curves demonstrated superior survival in the combination group compared with each treatment alone (P = 0.02). Flow cytometry showed increased infiltration of immunosuppressive cells as well as CTL in the radiation and combination groups (P = 0.04). Ratio of immunosuppressive cells to CTL shifted in favor of cytotoxic activity in the combination arm (P < 0.001). The qPCR analysis revealed downregulation of immunosuppressive genes (CCL22, IL22, and IL13), as well as upregulation of markers of CTL activation (CXCL9, GZMA, and GZMB) within both the radiated and distant tumors within the combination group. Combining radiation with immune checkpoint inhibitor provided better response in the radiated tumors and also the distant tumors along with a shift within the tumor microenvironment favoring cytotoxic activity. These findings demonstrate a possible abscopal effect in urothelial carcinoma with combination therapy.
Collapse
Affiliation(s)
- Alexis Rompré-Brodeur
- Urologic Oncology Research Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.,Department of Urology, McGill University Health Center, Montreal, Quebec, Canada
| | - Surashri Shinde-Jadhav
- Urologic Oncology Research Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada
| | - Mina Ayoub
- Urologic Oncology Research Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.,Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Ciriaco A Piccirillo
- Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Microbiology and Immunology, McGill University Health Center, Montreal, Quebec, Canada.,Program in Infectious Diseases and Immunology in Global Health, Centre for Translational Biology, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jan Seuntjens
- Department of Medical Physics, McGill University Health Center, Montreal, Quebec, Canada
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada
| | - Jose Joao Mansure
- Urologic Oncology Research Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada.,Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Wassim Kassouf
- Urologic Oncology Research Program, Research Institute of the McGill University Health Center, Montreal, Quebec, Canada. .,Department of Urology, McGill University Health Center, Montreal, Quebec, Canada.,Centre of Excellence in Translational Immunology (CETI), Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
44
|
Abstract
BCG immunotherapy is the gold-standard treatment for non-muscle-invasive bladder cancer at high risk of recurrence or progression. Preclinical and clinical studies have revealed that a robust inflammatory response to BCG involves several steps: attachment of BCG; internalization of BCG into resident immune cells, normal cells, and tumour urothelial cells; BCG-mediated induction of innate immunity, which is orchestrated by a cellular and cytokine milieu; and BCG-mediated initiation of tumour-specific immunity. As an added layer of complexity, variation between clinical BCG strains might influence development of tumour immunity. However, more than 40 years after the first use of BCG for bladder cancer, many questions regarding its mechanism of action remain unanswered. Clearly, a better understanding of the mechanisms underlying BCG-mediated tumour immunity could lead to improved efficacy, increased tolerance of treatment, and identification of novel immune-based therapies. Indeed, enthusiasm for bladder cancer immunotherapy, and the possibility of combining BCG with other therapies, is increasing owing to the availability of targeted immunotherapies, including checkpoint inhibitors. Understanding of the mechanism of action of BCG immunotherapy has advanced greatly, but many questions remain, and further basic and clinical research efforts are needed to develop new treatment strategies for patients with bladder cancer.
Collapse
|
45
|
Jagoda EM, Vasalatiy O, Basuli F, Opina ACL, Williams MR, Wong K, Lane KC, Adler S, Ton AT, Szajek LP, Xu B, Butcher D, Edmondson EF, Swenson RE, Greiner J, Gulley J, Eary J, Choyke PL. Immuno-PET Imaging of the Programmed Cell Death-1 Ligand (PD-L1) Using a Zirconium-89 Labeled Therapeutic Antibody, Avelumab. Mol Imaging 2019; 18:1536012119829986. [PMID: 31044647 PMCID: PMC6498777 DOI: 10.1177/1536012119829986] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The goal is to evaluate avelumab, an anti-PD-L1 monoclonal immunoglobulin G antibody labeled with zirconium-89 in human PD-L1-expressing cancer cells and mouse xenografts for clinical translation. METHODS [89Zr]Zr-DFO-PD-L1 monoclonal antibody (mAb) was synthesized using avelumab conjugated to desferrioxamine. In vitro binding studies and biodistribution studies were performed with PD-L1+MDA-MB231 cells and MDA-MB231 xenograft mouse models, respectively. Biodistributions were determined at 1, 2, 3, 5, and 7 days post coinjection of [89Zr]Zr-DFO-PD-L1 mAb without or with unlabeled avelumab (10, 20, 40, and 400 µg). RESULTS [89Zr]Zr-DFO-PD-L1 mAb exhibited high affinity (Kd ∼ 0.3 nM) and detected moderate PD-L1 expression levels in MDA-MB231 cells. The spleen and lymph nodes exhibited the highest [89Zr]Zr-DFO-PD-L1 mAb uptakes in all time points, while MDA-MB231 tumor uptakes were lower but highly retained. In the unlabeled avelumab dose escalation studies, spleen tissue-muscle ratios decreased in a dose-dependent manner indicating specific [89Zr]Zr-DFO-PD-L1 mAb binding to PD-L1. In contrast, lymph node and tumor tissue-muscle ratios increased 4- to 5-fold at 20 and 40 µg avelumab doses. CONCLUSIONS [89Zr]Zr-DFO-PD-L1 mAb exhibited specific and high affinity for PD-L1 in vitro and had target tissue uptakes correlating with PD-L1 expression levels in vivo. [89Zr]Zr-DFO-PD-L1 mAb uptake in PD-L1+tumors increased with escalating doses of avelumab.
Collapse
Affiliation(s)
- Elaine M Jagoda
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Olga Vasalatiy
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Falguni Basuli
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Ana Christina L Opina
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Mark R Williams
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Karen Wong
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Kelly C Lane
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Steve Adler
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Anita Thein Ton
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | | | - Biying Xu
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - Donna Butcher
- 4 Pathology & Histotechnology Lab Frederick National Laboratory for Cancer Research, NCI, Frederick, MD, USA
| | - Elijah F Edmondson
- 4 Pathology & Histotechnology Lab Frederick National Laboratory for Cancer Research, NCI, Frederick, MD, USA
| | - Rolf E Swenson
- 2 Imaging Probe Development Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, USA
| | - John Greiner
- 5 Laboratory of Tumor Immunology and Biology, National Cancer Institute, Bethesda, MD, USA
| | - James Gulley
- 6 Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD, USA.,7 Clinical Research Directorate/CMRP, Leidos Biomedical Research Inc. (formerly SAIC-Frederick, Inc.), Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Janet Eary
- 8 Cancer Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Peter L Choyke
- 1 Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
46
|
Zhang Y, Fang C, Wang RE, Wang Y, Guo H, Guo C, Zhao L, Li S, Li X, Schultz PG, Cao YJ, Wang F. A tumor-targeted immune checkpoint blocker. Proc Natl Acad Sci U S A 2019; 116:15889-15894. [PMID: 31332018 PMCID: PMC6689898 DOI: 10.1073/pnas.1905646116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To direct checkpoint inhibition to the tumor microenvironment, while avoiding systemic immune activation, we have synthesized a bispecific antibody [norleucine4, d-Phe7]-melanocyte stimulating hormone (NDP-MSH)-antiprogrammed cell death-ligand 1 antibody (αPD-L1) by conjugating a melanocyte stimulating hormone (α-MSH) analog to the antiprogrammed cell death-ligand 1 to (αPD-L1) antibody avelumab. This bispecific antibody can bind to both the melanocortin-1 receptor (MC1R) and to PD-L1 expressed on melanoma cells and shows enhanced specific antitumor efficacy in a syngeneic B16-SIY melanoma mouse model compared with the parental antibody at a 5 mg/kg dose. Moreover, the bispecific antibody showed increased infiltrated T cells in the tumor microenvironment. These results suggest that a tumor-targeted PD-L1-blocking bispecific antibody could have a therapeutic advantage in vivo, especially when used in combination with other checkpoint inhibitors.
Collapse
Affiliation(s)
- Yuhan Zhang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China
| | - Changming Fang
- California Institute for Biomedical Research (Calibr), La Jolla, CA 92037
| | - Rongsheng E Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Ying Wang
- California Institute for Biomedical Research (Calibr), La Jolla, CA 92037
| | - Hui Guo
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China
| | - Chao Guo
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China
- School of Ocean, Shandong University, 264209 Weihai, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, 518055 Shenzhen, China
| | - Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, 518055 Shenzhen, China
| | - Xia Li
- School of Ocean, Shandong University, 264209 Weihai, China
| | - Peter G Schultz
- California Institute for Biomedical Research (Calibr), La Jolla, CA 92037;
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, 518055 Shenzhen, China
| | - Feng Wang
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China;
| |
Collapse
|
47
|
Samishina EA, Blinova EV, Roshchin DA, Suslova IR, Blinov DS, Zhdanov PN, Deryabina ON, Kit'ko OV. Programmed death-ligand 1 signaling pathway involves in bladder cancer growth and progression. J Carcinog 2019; 18:3. [PMID: 31360124 PMCID: PMC6594057 DOI: 10.4103/jcar.jcar_3_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/09/2019] [Indexed: 01/12/2023] Open
Abstract
CONTEXT: Exploration of the biological property of programmed death-ligand 1 (PD-L1) signaling that may impact bladder tumor growth in humanized animals and cell culture. AIMS: The aim of this study is to evaluate how PD-L1 signaling involves bladder cancer growth and progression. SETTINGS AND DESIGN: This study design involves experimental in vivo and in vivo study. SUBJECTS AND METHODS: A role of PD-L1 signaling pathway inhibition for bladder cancer growth was assessed in humanized immunodeficient animals carried main molecular subtypes of bladder carcinoma patient-derived xenografts and provided with selective anti-PD-L1 treatment; bladder cancer cells invasiveness was evaluated in mixed RT112/84 cells + CD4+ cells culture incubated with PD-L1 blocker durvalumab. We used two-tailed Student's t-test to explore differences between main and control subgroups. Significance of intergroup comparison was measured with one-way ANOVA followed by the Tukey's or Newman–Keul's criterion. Survival curves were analyzed with Gehan's criterion with the Yate's correction. Differences were considered statistically significant at P < 0.05. RESULTS: Anti-PD-L1 intervention increased survival of the animals carried both primary and relapsed luminal noninvasive, muscular invasive, and relapsed luminal bladder cancer xenografts. There was significant retardation of tumor volume duplication time in aforementioned subgroups correlated with PD-L1 expression. Durvalumab treatment in concentration-dependent manner inhibited tumor cells invasiveness of mixed RT112 + CD4+ culture cells with its maximum at the highest studied concentration (10 μM). CONCLUSIONS: Obtained data constituted the pivotal role of programmed cell death-1/PD-L1 signaling pathway in bladder cancer development and progression. The results will have major implications for further clinical investigations.
Collapse
Affiliation(s)
- Elena A Samishina
- Department of General Pathology, Laboratory of Pharmacology, All-Union Scientific Center for Biological Active Compounds Safety, Staraja Kupavna, Russia
| | - Ekaterina V Blinova
- Department of Clinical Anatomy, Sechenov University, Moscow, Russia.,Department of Pharmacology, Laboratory of Molecular Pharmacology, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Dmitry A Roshchin
- Department of Oncological Urology, Russian National Research Medical Centre of Radiology, Moscow, Russia
| | - Irina R Suslova
- Department of Clinical Anatomy, Sechenov University, Moscow, Russia
| | - Dmitry S Blinov
- Department of General Pathology, Laboratory of Pharmacology, All-Union Scientific Center for Biological Active Compounds Safety, Staraja Kupavna, Russia
| | - Pavel N Zhdanov
- Department of General Pathology, Laboratory of Pharmacology, All-Union Scientific Center for Biological Active Compounds Safety, Staraja Kupavna, Russia
| | - Olga N Deryabina
- Department of Pharmacology, Laboratory of Molecular Pharmacology, National Research Ogarev Mordovia State University, Saransk, Russia
| | - Olesia V Kit'ko
- Department of Clinical Anatomy, Sechenov University, Moscow, Russia
| |
Collapse
|
48
|
Song D, Powles T, Shi L, Zhang L, Ingersoll MA, Lu YJ. Bladder cancer, a unique model to understand cancer immunity and develop immunotherapy approaches. J Pathol 2019; 249:151-165. [PMID: 31102277 PMCID: PMC6790662 DOI: 10.1002/path.5306] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/02/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022]
Abstract
With the mechanistic understanding of immune checkpoints and success in checkpoint blockade using antibodies for the treatment of certain cancers, immunotherapy has become one of the hottest areas in cancer research, with promise of long‐lasting therapeutic effect. Currently, however, only a proportion of cancers have a good response to checkpoint inhibition immunotherapy. Better understanding of the cancer response and resistance mechanisms is essential to fully explore the potential of immunotherapy to cure the majority of cancers. Bladder cancer, one of the most common and aggressive malignant diseases, has been successfully treated both at early and advanced stages by different immunotherapeutic approaches, bacillus Calmette–Guérin (BCG) intravesical instillation and anti‐PD‐1/PD‐L1 immune checkpoint blockade, respectively. Therefore, it provides a good model to investigate cancer immune response mechanisms and to improve the efficiency of immunotherapy. Here, we review bladder cancer immunotherapy with equal weight on BCG and anti‐PD‐1/PD‐L1 therapies and demonstrate why and how bladder cancer can be used as a model to study the predictors and mechanisms of cancer immune response and shine light on further development of immunotherapy approaches and response predictive biomarkers to improve immunotherapy of bladder cancer and other malignancies. We review the success of BCG and anti‐PD‐1/PD‐L1 treatment of bladder cancer, the underlying mechanisms and the therapeutic response predictors, including the limits to our knowledge. We then highlight briefly the adaptation of immunotherapy approaches and predictors developed in other cancers for bladder cancer therapy. Finally, we explore the potential of using bladder cancer as a model to investigate cancer immune response mechanisms and new therapeutic approaches, which may be translated into immunotherapy of other human cancers. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Dongkui Song
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Thomas Powles
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, UK.,Department of Medical Oncology, Barts Health NHS, London, UK
| | - Lei Shi
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Molly A Ingersoll
- Department of Immunology, Institut Pasteur, Paris, France.,Inserm U1223, Paris, France
| | - Yong-Jie Lu
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou University, Zhengzhou, PR China.,Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
49
|
Morillon YM, Su Z, Schlom J, Greiner JW. Temporal changes within the (bladder) tumor microenvironment that accompany the therapeutic effects of the immunocytokine NHS-IL12. J Immunother Cancer 2019; 7:150. [PMID: 31186063 PMCID: PMC6558846 DOI: 10.1186/s40425-019-0620-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/16/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND While significant strides in the treatment of metastatic bladder cancer have been made with immune checkpoint inhibitors, the treatment of carcinoma in situ and non-muscle invasive, non-metastatic (superficial) human urothelial carcinoma, also termed non-muscle invasive bladder cancer (NMIBC), remains intractable with bacillus Calmette-Guerin (BCG) employed as the standard of care. In this study, an immunocytokine, NHS-muIL12, which consists of two molecules of murine IL-12 fused to NHS76, a tumor necrosis-targeting human IgG1, was examined as an immunotherapeutic in an orthotopic MB49luc bladder tumor model. METHODS The antitumor activity of systemic administration of NHS-muIL12 was investigated on MB49luc tumors, an aggressive, bioluminescent orthotopic bladder cancer model. Temporal studies were carried out on MB49luc bladder tumors harvested during various time points during NHS-muIL12 treatment and cellular changes associated with the reduction in tumor burden following NHS-muIL12 were determined by flow cytometry. Effects of those changes on the proliferation/activation of lymphoid cells were also determined. RESULTS Studies revealed a significant reduction in MB49luc bladder tumor burden occurring between days 3 and 6 after the third and final systemic administration of NHS-muIL12. Temporal analyses of the MB49luc bladder tumor microenvironment (TME) initially revealed a large accumulation of myeloid-derived suppressor cells (MDSCs) and macrophages that elicited potent immunosuppression. Immunosuppression was characterized by the inability of CD4+ and CD8+ T cells to respond to broad-based immune stimulants. NHS-muIL12 administration resulted in temporal-dependent reductions in the number of MDSCs, macrophages and tumor-associated TGF-β, which culminated in a re-ignition of CD4+ and CD8+ T cells to elicit potent antitumor responses against MB49luc bladder tumors. CONCLUSIONS These findings provide strong evidence that the systemic administration of an immunocytokine consisting of a tumor-targeting Ig through recognition of DNA and DNA-histone complexes coupled to muIL-12 can effectively target the bladder TME; this significantly reduces the myeloid cellular compartment and reverts an immunosuppressive to an immunopermissive TME, ultimately resulting in antitumor effects. These studies provide further rationale for the employment of NHS-IL12 as an immunomodulator and clinical immunotherapeutic for NMIBC.
Collapse
Affiliation(s)
- Y Maurice Morillon
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Zhen Su
- EMD Serono, Rockland, MA, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
50
|
Blinova E, Roshchin D, Kogan E, Samishina E, Demura T, Deryabina O, Suslova I, Blinov D, Zhdanov P, Osmanov U, Nelipa M, Kaprin A. Patient-Derived Non-Muscular Invasive Bladder Cancer Xenografts of Main Molecular Subtypes of the Tumor for Anti-Pd-l1 Treatment Assessment. Cells 2019; 8:cells8060526. [PMID: 31159302 PMCID: PMC6628037 DOI: 10.3390/cells8060526] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/20/2019] [Accepted: 05/29/2019] [Indexed: 01/09/2023] Open
Abstract
Background: Establishment of heterotopic patient-derived xenografts of primary and relapsed non-muscular invasive bladder cancer (NMIBC) to explore the biological property of PD-L1 signaling that may impact bladder tumor growth in humanized animals. Methods: Tumor cells of luminal, basal, and p53 subtypes of primary and relapsed NMIBC were engrafted to irradiated (3.5 Gy) NOG/SCID female mice along with intraperitoneal transplantation of human lymphocytes (5 × 107 cells/mouse); a role of PD-L1 signaling pathway inhibition for bladder cancer growth was assessed in humanized animals that carried PD-L1-expressing main molecular subtypes of bladder carcinoma patient-derived xenografts (PDX) and provided with selective anti-PD-L1 treatment. We used two-tailed Student’s t test to explore differences between main and control subgroups. Significance of intergroup comparison was measured with one-way ANOVA followed by the Tukey’s or Newman–Keul’s criterion. Survival curves were analyzed with the Gehan’s criterion with the Yate’s correction. The Spearman’s correlation was used to assess the link between CD8+ expression and sPD-L1 serum level. Differences were considered statistically significant at p < 0.05. Results: Heterotopic primary and relapsed luminal, basal, and p53 subtypes of NMIBC PDXs were established. More than 25% of counted tumor cells of all PDX specimens expressed PD-L1, so the tumors were ranged as PD-L1 positive. Anti-PD-L1 intervention increased survival of the animals that carried both primary and relapsed luminal noninvasive, muscular invasive, and relapsed luminal bladder cancer xenografts. There was significant retardation of tumor volume duplication time in aforementioned subgroups correlated with PD-L1 expression. Bad response of p53 mutant subtypes of NMIBC on specific anti-PD-L1 treatment may be associated with low CD8+ cells representation into the tumors tissue. Conclusions: Established PD-L1-positive NMIBC PDXs differently replied on anti-PD-L1 treatment due to both NMIBC molecular subtype and tumor T-suppressors population. The results may have major implications for further clinical investigations.
Collapse
Affiliation(s)
- Ekaterina Blinova
- Department of Pathology; Department of Operative Surgery and Clinical Anatomy, Sechenov University, 8/1 Trubetzkaya Street, 119991 Moscow, Russia.
- Department of Oncology; Laboratory of Pharmacology, National Research Ogarev Mordovia State University, 68 Bolshevistskaya Street, 430005 Saransk, Russia.
| | - Dmitry Roshchin
- Department of Oncological urology, Russian National Research Medical Center of Radiology, 3 2nd Botkinsky Proezd, 125284 Moscow, Russia.
| | - Evgenya Kogan
- Department of Pathology; Department of Operative Surgery and Clinical Anatomy, Sechenov University, 8/1 Trubetzkaya Street, 119991 Moscow, Russia.
| | - Elena Samishina
- Laboratory of Molecular Pharmacology, All-Union Research Center for Biological Active Compounds Safety, 23 Kirova Street, 142450 Staraja Kupavna, Russia.
| | - Tatiana Demura
- Department of Pathology; Department of Operative Surgery and Clinical Anatomy, Sechenov University, 8/1 Trubetzkaya Street, 119991 Moscow, Russia.
| | - Olga Deryabina
- Department of Oncology; Laboratory of Pharmacology, National Research Ogarev Mordovia State University, 68 Bolshevistskaya Street, 430005 Saransk, Russia.
| | - Irina Suslova
- Laboratory of Molecular Pharmacology, All-Union Research Center for Biological Active Compounds Safety, 23 Kirova Street, 142450 Staraja Kupavna, Russia.
| | - Dmitry Blinov
- Laboratory of Molecular Pharmacology, All-Union Research Center for Biological Active Compounds Safety, 23 Kirova Street, 142450 Staraja Kupavna, Russia.
| | - Pavel Zhdanov
- Laboratory of Molecular Pharmacology, All-Union Research Center for Biological Active Compounds Safety, 23 Kirova Street, 142450 Staraja Kupavna, Russia.
| | - Usif Osmanov
- Department of Pathology; Department of Operative Surgery and Clinical Anatomy, Sechenov University, 8/1 Trubetzkaya Street, 119991 Moscow, Russia.
| | - Mikhail Nelipa
- Department of Pathology; Department of Operative Surgery and Clinical Anatomy, Sechenov University, 8/1 Trubetzkaya Street, 119991 Moscow, Russia.
| | - Andrey Kaprin
- Department of Oncological urology, Russian National Research Medical Center of Radiology, 3 2nd Botkinsky Proezd, 125284 Moscow, Russia.
| |
Collapse
|