1
|
Liu J, Yu Y, Liu C, Gao C, Zhuang J, Liu L, Wu Q, Ma W, Zhang Q, Sun C. Combinatorial regimens of chemotherapeutic agents: A new perspective on raising the heat of the tumor immune microenvironment. Front Pharmacol 2022; 13:1035954. [PMID: 36304169 PMCID: PMC9593050 DOI: 10.3389/fphar.2022.1035954] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Harnessing the broad immunostimulatory capabilities of chemotherapy in combination with immune checkpoint inhibitors has improved immunotherapy outcomes in patients with cancer. Certain chemotherapeutic agents can extensively modify the tumor microenvironment (TME), resulting in the reprogramming of local immune responses. Although chemotherapeutic agents with an enhanced generation of potent anti-tumor immune responses have been tested in preclinical animal models and clinical trials, this strategy has not yet shown substantial therapeutic efficacy in selected difficult-to-treat cancer types. In addition, the efficacy of chemotherapeutic agent-based monotherapy in eliciting a long-term anti-tumor immune response is restricted by the immunosuppressive TME. To enhance the immunomodulatory effect of chemotherapy, researchers have made many attempts, mainly focusing on improving the targeted distribution of chemotherapeutic agents and designing combination therapies. Here, we focused on the mechanisms of the anti-tumor immune response to chemotherapeutic agents and enumerated the attempts to advance the use of chemo-immunotherapy. Furthermore, we have listed the important considerations in designing combinations of these drugs to maximize efficacy and improve treatment response rates in patients with cancer.
Collapse
Affiliation(s)
- Jingyang Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yang Yu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Qiming Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Qiming Zhang, ; Changgang Sun,
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
- *Correspondence: Qiming Zhang, ; Changgang Sun,
| |
Collapse
|
2
|
Principe DR, Kamath SD, Korc M, Munshi HG. The immune modifying effects of chemotherapy and advances in chemo-immunotherapy. Pharmacol Ther 2022; 236:108111. [PMID: 35016920 PMCID: PMC9271143 DOI: 10.1016/j.pharmthera.2022.108111] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/06/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment paradigm for several malignancies. While the use of single-agent or combined ICIs has achieved acceptable disease control rates in a variety of solid tumors, such approaches have yet to show substantial therapeutic efficacy in select difficult-to-treat cancer types. Recently, select chemotherapy regimens are emerging as extensive modifiers of the tumor microenvironment, leading to the reprogramming of local immune responses. Accordingly, data is now emerging to suggest that certain anti-neoplastic agents modulate various immune cell processes, most notably the cross-presentation of tumor antigens, leukocyte trafficking, and cytokine biosynthesis. As such, the combination of ICIs and cytotoxic chemotherapy are beginning to show promise in many cancers that have long been considered poorly responsive to ICI-based immunotherapy. Here, we discuss past and present attempts to advance chemo-immunotherapy in these difficult-to-treat cancer histologies, mechanisms through which select chemotherapies modify tumor immunogenicity, as well as important considerations when designing such approaches to maximize efficacy and improve therapeutic response rates.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA; Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Suneel D Kamath
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Murray Korc
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Hidayatullah G Munshi
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
3
|
Anacardic 6-pentadecyl salicylic acid induces apoptosis in breast cancer tumor cells, immunostimulation in the host and decreases blood toxic effects of taxol in an animal model. Toxicol Appl Pharmacol 2020; 410:115359. [PMID: 33290779 DOI: 10.1016/j.taap.2020.115359] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 01/12/2023]
Abstract
Many antineoplastic agents induce myelosuppression and leukopenia as secondary effects in patients. The development of anticancer agents that simultaneously provoke antitumor immune response represents an important therapeutic advance. The administration of 6-pentadecyl salicylic acid (6SA) contributes to the antitumor immunity using 4T1 breast cancer cells in Balb/c female mice, with Taxol as a positive control and in cotreatment with 6SA (6SA + Taxol; CoT). Our results show that 6SA reduces tumor volume and size by inducing caspase-8-mediated apoptosis without reducing tumor infiltrated lymphocytes. Also, 6SA reduced lung metastasis and increased the proportion of immune cells in blood, lymph nodes and bone marrow; more evidently, in the proportion of tumor-infiltrated natural killer (NK) cells and cytotoxic T lymphocytes. Taxol reduces helper and cytotoxic lymphocytes causing systemic immunosuppression and myelosuppression in bone marrow, whereas 6SA does not decrease any immune cell subpopulations in circulating blood and lymph nodes. More importantly, the CoT decreased the Taxol-induced cytotoxicity in circulating T cells and bone marrow. Treatment with 6SA increases the secretion of IL-2, IL-12, GM-CSF, TNF-α and IFN-γ and significantly reduces IL-10 and IL-17 secretion, suggesting that the reduction of regulatory T cells and tumor-associated macrophages contribute to the host control of tumor development. Finally, 6SA has an effective antineoplastic activity against breast cancer cells in an immunocompetent animal, reduces the myelosuppression and leukopenia that Taxol produces, improves the antitumoral immunological microenvironment and increases the overall survival of the animals improving the quality of life of patients with cancer.
Collapse
|
4
|
Bose C, Banerjee P, Kundu J, Dutta B, Ghosh I, Sinha S, Ghosh A, Barua A, Gupta S, Das U, Jana SS, Sinha S. Evaluation of a Tubulin‐Targeted Pyrimidine Indole Hybrid Molecule as an Anticancer Agent. ChemistrySelect 2020. [DOI: 10.1002/slct.202003322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chandra Bose
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Priyanjalee Banerjee
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Jayanta Kundu
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Biswadeb Dutta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Indranil Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Shreya Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Argha Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Abhishek Barua
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Shalini Gupta
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Ujjal Das
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| | - Siddhartha S. Jana
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032 India
| | - Surajit Sinha
- School of Applied and Interdisciplinary Sciences, Indian Association for the Cultivation of Science, Jadavpur Kolkata 700032
| |
Collapse
|
5
|
Wang X, Xu W, Chi Z, Si L, Sheng X, Kong Y, Zhou L, Mao L, Lian B, Tang B, Yan X, Bai X, Cui C, Guo J. Chemotherapy combined with antiangiogenic drugs as salvage therapy in advanced melanoma patients progressing on PD-1 immunotherapy. Transl Oncol 2020; 14:100949. [PMID: 33221685 PMCID: PMC7689327 DOI: 10.1016/j.tranon.2020.100949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/31/2020] [Accepted: 11/05/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND This study aimed to evaluate the effect of salvage therapy with nab-paclitaxel (nab-p) or temozolomide (TMZ) combined with antiangiogenic drugs in programmed death 1 (PD-1) inhibitor-resistant patients with unresectable metastatic melanoma. METHODS We conducted a retrospective review of 69 metastatic melanoma patients who received nab-p or TMZ combined with antiangiogenic drugs after developing PD-1 inhibitor resistance and were treated at the Beijing Cancer Hospital between 2016 and 2019. The disease control rate (c-DCR) and progression-free survival (c-PFS) of salvage CA (chemotherapy combined with antiangiogenic drugs) regimens were investigated. Univariate and multivariate analyses were performed to evaluate the clinical pathological factors affecting the outcomes. Then, a nomogram was formulated to predict the probability of 3-month and 6-month c-PFS based on the multivariate analysis results. RESULTS The c-DCR was 63.8%, and the median c-PFS was 3.0 months. In the univariate analysis, factors associated with the c-DCR were included the melanoma subtype, baseline platelet-to-lymphocyte ratio (PLR) and best response status to PD-1 inhibitors. Factors influencing c-PFS included age, baseline lactic dehydrogenase, PLR, neutrophil-to-lymphocyte ratio (NLR), PFS duration of anti-PD-1 therapy (p-PFS), and the best response and progression pattern of PD-1 inhibitors. In the multivariate analysis, age <65 years, heterogeneous progression pattern and baseline PLR<200 were significantly associated with improved c-PFS. The concordance index (C-index) of the nomogram was equal to 0.65 (95% CI 0.566-0.734). CONCLUSIONS CA regimens demonstrated promising effects in PD-1 inhibitor-resistant patients. The nomogram could be a valuable predictive module for salvage therapy choice in PD-1 inhibitor-resistant patients.
Collapse
Affiliation(s)
- Xuan Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Weiran Xu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Zhihong Chi
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lu Si
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xinan Sheng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Yan Kong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Li Zhou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Lili Mao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Bin Lian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Bixia Tang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xieqiao Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Xue Bai
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Chuanliang Cui
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China
| | - Jun Guo
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Haidian District, Beijing 100142, China.
| |
Collapse
|
6
|
Guo CH, Hsia S, Chung CH, Lin YC, Shih MY, Chen PC, Peng CL, Henning SM, Hsu GSW, Li Z. Nutritional supplements in combination with chemotherapy or targeted therapy reduces tumor progression in mice bearing triple-negative breast cancer. J Nutr Biochem 2020; 87:108504. [PMID: 32956826 DOI: 10.1016/j.jnutbio.2020.108504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 08/26/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
The potential anti-cancer properties of selenium (Se) and eicosapentaenoic acid (EPA)/docosahexaenoic acid (DHA) have been documented. However, few studies have been conducted examining anti-tumor effects of nutritional supplements (NS) containing Se and EPA/DHA in combination with anti-cancer agents, such as taxol (Tax), adriamycin (Adr), and avastin (Ava). Compared with triple-negative breast cancer (TNBC)-bearing positive control (TB) mice, a low dose of Tax, Adr, and Ava decreased tumor size and the incidence of metastasis in TB-Tax, TB-Adr, and TB-Ava groups. Combination treatment with anti-cancer agent and NS (2.7 μg Se and 5.1 mg EPA/3.7 mg DHA/g) induced additional decreases in TB-Tax-NS, TB-Adr-NS, and TB-Ava-NS groups. Th1-associated cytokines were increased, and Th2-type cytokines were decreased significantly in TB mice with combination treatment than that of anti-cancer agent treatment alone. Combination treatment with anti-cancer agents and NS has also been shown to further increased tumor malondialdehyde (MDA) levels, lowered hypoxia-inducible factor (HIF)-1α, angiogenic markers (vascular endothelial growth factor [VEGF] and CD31) and metastatic potential, as well as reduced heat shock proteins, receptor tyrosine kinase AXL, and surface markers of cancer stem cells, and increased apoptotic proteins. For immune checkpoint molecules, combination treatment was associated with a greater decrease in programmed cell death ligand-1 (PD-L1) in both tumors and mammary glands, but PD-1 level in primary tumors was increased. Our results suggest that combination treatment with low-dose anti-cancer agents (Tax, Adr, and Ava) and oral supplementation of Se/ EPA/DHA significantly decreased tumor growth and metastatic progression in TNBC mice through multiple anti-tumor mechanisms.
Collapse
Affiliation(s)
- Chih-Hung Guo
- Micronutrition and Biomedical Nutrition Lab, Institute of Biomedical Nutrition, Hung-Kuang University, Taichung 433, Taiwan; Taiwan Nutraceutical Association, Taipei 105, Taiwan.
| | - Simon Hsia
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | | | - Yi-Chun Lin
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | - Min-Yi Shih
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | | | - Chia-Lin Peng
- Taiwan Nutraceutical Association, Taipei 105, Taiwan
| | - Susanne M Henning
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | - Guoo-Shyng W Hsu
- Department of Nutritional Science, Fu Jen University, New Taipei City 242, Taiwan
| | - Zhaoping Li
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| |
Collapse
|
7
|
Zhao L, Ma B, Yang Y, Li T, Han L, Gao Q. Chemotherapy Reverses Anti-PD-1 Resistance in One Patient With Advanced Non-small Lung Cell Cancer. Front Oncol 2020; 10:507. [PMID: 32373522 PMCID: PMC7186846 DOI: 10.3389/fonc.2020.00507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 03/20/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Programmed cell death protein 1(PD-1) blockade has become a standard second-line treatment option for patients with advanced non-small cell lung cancer (NSCLC) without a driver gene mutation. Previous clinical studies showed that the objective response rate (ORR) of PD-1 blockade as second-line treatment for patients with NSCLC was ~20%, and the median progression-free survival (PFS) was ~4 months, with most patients eventually developing a resistance to PD-1 blockade. Although the ORR to chemotherapy after PD-1 blockade resistance was relatively high, the survival time of patients could not be significantly prolonged. Clinical oncologists are unclear about which treatment regimen should be selected after PD-1 blockade failure. Here, we report about a patient with advanced NSCLC and initial PD-1 blockade resistance who was observed to have a rapid partial response (PR) following one dose of chemotherapy and subsequent PD-1 blockade treatment. Case presentation: A 70-year-old woman with a history of left lower lobe lung surgery in March 2018 (pathological stage T1N2M0, EGFR wild-type) presented to our hospital. After six cycles of adjuvant chemotherapy, multiple nodules in both the lungs developed, and were suspected to be metastatic lesions. After another 2 months, the nodules in both the lungs enlarged. From November 2018 to March 2019, the patient received six cycles of pembrolizumab, and computed tomography (CT) confirmed a progressive disease status. She was then managed with 260 mg/m2 albumin paclitaxel once every 3 weeks. Subsequently, chemotherapy was discontinued after one cycle owing to grade three neuromuscular toxicity. Follow-up CT revealed a stable disease in May 2019. She then received another six cycles of pembrolizumab, which resulted in a PR. Conclusion: Chemotherapy may play a role in reversing PD-1 blockade resistance. If failure of PD-1 blockade occurs at first, re-administration of PD-1 blockade may be implemented if first followed by several cycles of chemotherapy. Because there are few reports on the use of chemotherapy to reverse PD-1 resistance, it is necessary to conduct clinical studies with larger patient cohorts to investigate whether chemotherapy can reverse PD-1 blockade resistance.
Collapse
Affiliation(s)
- Lingdi Zhao
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Baozhen Ma
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonghao Yang
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Tiepeng Li
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Han
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Quanli Gao
- Department of Immunotherapy, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305:130-154. [DOI: 10.1016/j.jconrel.2019.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
|
9
|
Abstract
Although common evolutionary principles drive the growth of cancer cells regardless of the tissue of origin, the microenvironment in which tumours arise substantially differs across various organ sites. Recent studies have established that, in addition to cell-intrinsic effects, tumour growth regulation also depends on local cues driven by tissue environmental factors. In this Review, we discuss how tissue-specific determinants might influence tumour development and argue that unravelling the tissue-specific contribution to tumour immunity should help the development of precise immunotherapeutic strategies for patients with cancer.
Collapse
Affiliation(s)
- Hélène Salmon
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- INSERM U932, Institut Curie, Paris, France.
| | | | - Sacha Gnjatic
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
10
|
Soliman HH. nab-Paclitaxel as a potential partner with checkpoint inhibitors in solid tumors. Onco Targets Ther 2016; 10:101-112. [PMID: 28053544 PMCID: PMC5189972 DOI: 10.2147/ott.s122974] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumors recognized by the host immune system are associated with better survival. However, the immune system is often suppressed in patients with established tumor burden. Stimulating the immune system to detect and kill tumor cells has been a challenge in cancer therapy for some time. Recently, novel cancer immunotherapies, such as immune checkpoint inhibitors, monoclonal antibodies, and vaccine therapies, have emerged as promising therapeutic approaches for many solid tumors. However, for some tumors, immunotherapy alone has not provided significant benefits, and some may even be fully resistant to immunotherapy. It has been suggested that the immune system may require "priming" before an immunotherapy can elicit an immune response. Although chemotherapies are believed to be immunosuppressive, when given at the right dose and sequence these agents may provide this "priming" effect for the immune system. In addition to direct cytotoxic killing of tumor cells, standard chemotherapeutic agents can elicit immunogenicity through various mechanisms. This review highlights the general immunomodulatory properties of chemotherapy agents. It also provides a rationale for combined therapy with nab-paclitaxel and immune checkpoint inhibitors. Recent clinical trial data with these combination regimens in solid tumors are presented, along with a summary of ongoing trials.
Collapse
Affiliation(s)
- Hatem H Soliman
- Department of Oncologic Sciences, Moffitt Cancer Center and Research Institute, University of South Florida, Tampa, FL, USA
| |
Collapse
|
11
|
Remark R, Lupo A, Alifano M, Biton J, Ouakrim H, Stefani A, Cremer I, Goc J, Régnard JF, Dieu-Nosjean MC, Damotte D. Immune contexture and histological response after neoadjuvant chemotherapy predict clinical outcome of lung cancer patients. Oncoimmunology 2016; 5:e1255394. [PMID: 28123901 DOI: 10.1080/2162402x.2016.1255394] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/25/2016] [Accepted: 10/26/2016] [Indexed: 12/17/2022] Open
Abstract
There is now growing evidence that the immune contexture influences cancer progression and clinical outcome of patients with non-small cell lung cancer (NSCLC). If chemotherapy is widely used to treat patients with advanced-stage NSCLC, it remains unclear how it could modify the immune contexture and impact its prognostic value. Here, we analyzed two retrospective cohorts, respectively composed of 122 stage III-N2 NSCLC patients treated with chemotherapy before surgery and 39 stage-matched patients treated by surgery only. In patients treated with neoadjuvant chemotherapy, the histological characteristics, the expression of PD-L1 protein, and the tumor immune microenvironment (CD8+ T cells, DC-LAMP+ mature dendritic cells, and CD68+ macrophages) were evaluated and their prognostic value assessed together with standard clinical parameters. By analyzing pre- and post-treatment specimens, we did not find any changes in the PD-L1 expression. We also found that the tumor immune contexture in patients treated with neoadjuvant chemotherapy exhibited a similar pattern that the one found in chemotherapy-naive patients, with comparable densities of tumor-infiltrating CD8+ and DC-LAMP+ cells and a similar spatial organization. The percentage of residual viable tumor cells and the immune pattern (CD8+ and DC-LAMP+ cell densities) were significantly associated with the clinical outcome and allowed the identification of short- and long-term survivors, respectively. In multivariate analysis, the immune pattern was found to be the strongest independent prognostic factor. In conclusion, this study decrypts the complex interplay between cancer and immune cells in patients undergoing chemotherapy and supports potential beneficial synergistic effect of immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Romain Remark
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France; Denis Diderot-Paris 7 University, Paris, France
| | - Audrey Lupo
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France; Denis Diderot-Paris 7 University, Paris, France; Pathology Department, Cochin hospital, AP-HP, Paris, France
| | - Marco Alifano
- Paris Descartes-Paris 5 University, Paris, France; Thoracic Surgery Department, Cochin hospital, AP-HP, Paris, France
| | - Jerome Biton
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France
| | - Hanane Ouakrim
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France; Pathology Department, Cochin hospital, AP-HP, Paris, France
| | | | - Isabelle Cremer
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France
| | - Jeremy Goc
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France
| | - Jean-Francois Régnard
- Paris Descartes-Paris 5 University, Paris, France; Thoracic Surgery Department, Cochin hospital, AP-HP, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France
| | - Diane Damotte
- INSERM U1138, Team "Cancer, Immune Control, and Escape" Cordeliers Research Center, Paris, France; Pierre et Marie Curie-Paris 6 University, Paris, France; Paris Descartes-Paris 5 University, Paris, France; Pathology Department, Cochin hospital, AP-HP, Paris, France
| |
Collapse
|
12
|
Diederich M, Cerella C. Non-canonical programmed cell death mechanisms triggered by natural compounds. Semin Cancer Biol 2016; 40-41:4-34. [PMID: 27262793 DOI: 10.1016/j.semcancer.2016.06.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022]
Abstract
Natural compounds are the fundament of pharmacological treatments and more than 50% of all anticancer drugs are of natural origins or at least derived from scaffolds present in Nature. Over the last 25 years, molecular mechanisms triggered by natural anticancer compounds were investigated. Emerging research showed that molecules of natural origins are useful for both preventive and therapeutic purposes by targeting essential hallmarks and enabling characteristics described by Hanahan and Weinberg. Moreover, natural compounds were able to change the differentiation status of selected cell types. One of the earliest response of cells treated by pharmacologically active compounds is the change of its morphology leading to ultra-structural perturbations: changes in membrane composition, cytoskeleton integrity, alterations of the endoplasmic reticulum, mitochondria and of the nucleus lead to formation of morphological alterations that are a characteristic of both compound and cancer type preceding cell death. Apoptosis and autophagy were traditionally considered as the most prominent cell death or cell death-related mechanisms. By now multiple other cell death modalities were described and most likely involved in response to chemotherapeutic treatment. It can be hypothesized that especially necrosis-related phenotypes triggered by various treatments or evolving from apoptotic or autophagic mechanisms, provide a more efficient therapeutic outcome depending on cancer type and genetic phenotype of the patient. In fact, the recent discovery of multiple regulated forms of necrosis and the initial elucidation of the corresponding cell signaling pathways appear nowadays as important tools to clarify the immunogenic potential of non-canonical forms of cell death induction.
Collapse
Affiliation(s)
- Marc Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul 151-742, South Korea.
| | - Claudia Cerella
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| |
Collapse
|
13
|
Chiang CLL, Balint K, Coukos G, Kandalaft LE. Potential approaches for more successful dendritic cell-based immunotherapy. Expert Opin Biol Ther 2015; 15:569-82. [DOI: 10.1517/14712598.2015.1000298] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14
|
Abstract
While therapeutic vaccines for ovarian cancer represent only a small fraction of active clinical trials, growing interest in this area and the accumulated data supporting the use of vaccines in cancer treatment portend further expansion of trials incorporating these strategies. This review explores the rationale for the use of vaccines for the treatment of ovarian cancer. It examines vaccine platforms that have been investigated and reviews the data from these studies. We also highlight recently reported phase 2 and 3 clinical trials with clinical outcomes as endpoints. Finally, we consider directions for the next generation of vaccines in light of these findings and our emerging understanding of agents that may augment vaccine responses by targeting the immunosuppressive impact of the tumor microenvironment.
Collapse
|
15
|
Wen CC, Chen HM, Yang NS. Developing Phytocompounds from Medicinal Plants as Immunomodulators. ADVANCES IN BOTANICAL RESEARCH 2012; 62:197-272. [PMID: 32300254 PMCID: PMC7150268 DOI: 10.1016/b978-0-12-394591-4.00004-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Imbalance or malfunction of the immune systems is associated with a range of chronic diseases including autoimmune diseases, allergies, cancers and others. Various innate and adaptive immune cells that are integrated in this complex networking system may represent promising targets for developing immunotherapeutics for treating specific immune diseases. A spectrum of phytochemicals have been isolated, characterized and modified for development and use as prevention or treatment of human diseases. Many cytotoxic drugs and antibiotics have been developed from phytocompounds, but the application of traditional or new medicinal plants for use as immunomodulators in treating immune diseases is still relatively limited. In this review, a selected group of medicinal herbs, their derived crude or fractionated phytoextracts and the specific phytochemicals/phytocompounds isolated from them, as well as categorized phytocompound groups with specific chemical structures are discussed in terms of their immunomodulatory bioactivities. We also assess their potential for future development as immunomodulatory or inflammation-regulatory therapeutics or agents. New experimental approaches for evaluating the immunomodulatory activities of candidate phytomedicines are also discussed.
Collapse
Affiliation(s)
- Chih-Chun Wen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Hui-Ming Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Ning-Sun Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
16
|
Wen CC, Chen HM, Chen SS, Huang LT, Chang WT, Wei WC, Chou LC, Arulselvan P, Wu JB, Kuo SC, Yang NS. Specific microtubule-depolymerizing agents augment efficacy of dendritic cell-based cancer vaccines. J Biomed Sci 2011; 18:44. [PMID: 21689407 PMCID: PMC3141632 DOI: 10.1186/1423-0127-18-44] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Accepted: 06/20/2011] [Indexed: 12/13/2022] Open
Abstract
Background Damage-associated molecular patterns (DAMPs) are associated with immunogenic cell death and have the ability to enhance maturation and antigen presentation of dendritic cells (DCs). Specific microtubule-depolymerizing agents (MDAs) such as colchicine have been shown to confer anti-cancer activity and also trigger activation of DCs. Methods In this study, we evaluated the ability of three MDAs (colchicine and two 2-phenyl-4-quinolone analogues) to induce immunogenic cell death in test tumor cells, activate DCs, and augment T-cell proliferation activity. These MDAs were further evaluated for use as an adjuvant in a tumor cell lysate-pulsed DC vaccine. Results The three test phytochemicals considerably increased the expression of DAMPs including HSP70, HSP90 and HMGB1, but had no effect on expression of calreticulin (CRT). DC vaccines pulsed with MDA-treated tumor cell lysates had a significant effect on tumor growth, showed cytotoxic T-lymphocyte activity against tumors, and increased the survival rate of test mice. In vivo antibody depletion experiments suggested that CD8+ and NK cells, but not CD4+ cells, were the main effector cells responsible for the observed anti-tumor activity. In addition, culture of DCs with GM-CSF and IL-4 during the pulsing and stimulation period significantly increased the production of IL-12 and decreased production of IL-10. MDAs also induced phenotypic maturation of DCs and augmented CD4+ and CD8+ T-cell proliferation when co-cultured with DCs. Conclusions Specific MDAs including the clinical drug, colchicine, can induce immunogenic cell death in tumor cells, and DCs pulsed with MDA-treated tumor cell lysates (TCLs) can generate potent anti-tumor immunity in mice. This approach may warrant future clinical evaluation as a cancer vaccine.
Collapse
Affiliation(s)
- Chih-Chun Wen
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Li Y, Efferson CL, Ramesh R, Peoples GE, Hwu P, Ioannides CG. A peptidoglycan monomer with the glutamine to serine change and basic peptides bind in silico to TLR-2 (403-455). Cancer Immunol Immunother 2011; 60:515-24. [PMID: 21188584 PMCID: PMC11028711 DOI: 10.1007/s00262-010-0959-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2010] [Accepted: 12/08/2010] [Indexed: 12/30/2022]
Abstract
Bacterial cell wall polysaccharides, such as PGN, bind and activate TLR-2, NOD2 and PGRP on monocytes/macrophages and activate inflammation. We found that the peptides containing basic amino acids (cations) at N -terminus and tyrosine at C-terminus interfered with activating ability of PGN. This finding is significant because the ECD of TLR-2 in vivo encounters a large number of proteins or peptides. Some should bind ECD and "pre-form" TLR-2 to respond or not to its activators, although they cannot activate TLR-2 alone. TLR-2 is receptor for a large number of ligands, including lipopeptides and bacterial cell wall glycoproteins. A binding site for lipopeptides has been identified; however, a binding site for soluble or multimeric PGN has not been proposed. To identify the candidate binding sites of peptides and PGN on TLR-2, we modeled docking of peptides and of the PGN monomer (PGN-S-monomer) to extracellular domain (ECD-TLR-2) of the unbound TLR-2. Quantification, in silico, of free energy of binding (DG) identified 2 close sites for peptides and PGN. The PGN-S-monomer binding site is between amino acids TLR-2, 404-430 or more closely TLR-2, 417-428. The peptide-binding site is between amino acids TLR-2, 434-455. Molecular models show PGN-S-monomer inserts its N -acetyl-glucosamine (NAG) deep in the TLR-2 coil, while its terminal lysine interacts with inside (Glu(403)) and outside pocket (Tyr(378)). Peptides insert their two N -terminal arginines or their C-terminal tyrosines in the TLR-2 coil. PGN did not bind the lipopeptide-binding site in the TLR-2. It can bind the C-terminus, 572-586 (DG = 0.026 kcal), of "lipopeptide-bound" TLR-2. An additional, low-affinity PGN-binding site is TLR-2 (227-237). MTP, MDP, and lysine-less PGN bind to TLR-2, 87-113. This is the first report identifying candidate binding sites of monomer PGN and peptides on TLR-2. Experimental verification of our findings is needed to create synthetic adjuvant for vaccines. Such synthetic PGN can direct both adjuvant and cancer antigen to TLR-2.
Collapse
Affiliation(s)
- Yufeng Li
- Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
- Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, P.O. Box 304, Houston, TX 77030 USA
| | - Clay L. Efferson
- Departments of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
- Present Address: Merck Corporation, Boston, MA USA
| | - Rajagopal Ramesh
- Departments of Thoracic Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - George E. Peoples
- Department of Surgery, General Surgery Service, Brooke Army Medical Center, 3851 Roger Brooke Drive, Fort Sam, Houston, TX 78234 USA
| | - Patrick Hwu
- Departments of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030 USA
| | - Constantin G. Ioannides
- Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, P.O. Box 304, Houston, TX 77030 USA
| |
Collapse
|
18
|
Mine T, Matsueda S, Gao H, Li Y, Wong KK, Peoples GE, Ferrone S, Ioannides CG. Created Gli-1 duplex short-RNA (i-Gli-RNA) eliminates CD44 Hi progenitors of taxol-resistant ovarian cancer cells. Oncol Rep 2010; 23:1537-43. [PMID: 20428807 DOI: 10.3892/or_00000793] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Notch and Hedgehog activate cell-cycle progression of adult and cancer stem cells. Notch is activated by DLL and Jag presents on neighboring cells. We investigated the effects of density of the Notch-activating ligand, Jag-1, and targeting Gli-1, in activation of division of paclitaxel/taxol-resistant, (PTX Res) ovarian cancer cells SKOV3 (SKOV3). We used the specific gamma-presenilin inhibitor, DAPT, to identify the specificity of activating signals for Notch-1 and created 'butterfly-duplex-3548-Gli-1-inhibitory RNA' (i-Gli-1.RNA) to inhibit cell division. To accurately quantify kinetics of division, the expression of CD44 and CD24 was determined in each gated population of divided cells. CD44 High proliferated when activated by Jag-1 Low and poorly when activated by Jag-1 High. DAPT inhibited proliferation of cells activated by Jag-1 Low, and increased proliferation of cells activated by Jag-1 High. Only 5-10% of cells activated by Jag-1 High and Jag-1 Low divided fast, polynomial, and symmetric. i-Gli-1.RNA eliminated more than 50% of the small CD44 High/CD24 Neg cells in divisions 3 and 4. This effect appeared specific compared with cells transfected with negative control siRNA. i-Gli-1.RNA had no effect on large CD44 High/CD24 Neg cells, but inhibited the population of CD44 High/CD24 Low cells. Expansion of CD44 High inversely correlated with Jag-1 density on activating autologous tumor and fibrosarcoma cells. Created i-RNAs may decrease the resting CSC pool. Notch and Gli-1 signals play an important role in proliferation/division and survival of cancer stem cells. Targeting Notch-1 through its enhancer Gl-1, should be significant for novel treatments to eliminate taxol-resistant cancer stem cells (CSC). i.Gli-1 RNA should be more effective if used together with Taxol.
Collapse
Affiliation(s)
- Takashi Mine
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Roy A, Singh MS, Upadhyay P, Bhaskar S. Combined chemo-immunotherapy as a prospective strategy to combat cancer: a nanoparticle based approach. Mol Pharm 2010; 7:1778-88. [PMID: 20822093 DOI: 10.1021/mp100153r] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The prime objective of this study was to develop a combined chemo-immunotherapeutic formulation which could directly kill cancer cells as well as activate the immunosuppressed tumor microenvironment to mount a robust antitumor immune response. Paclitaxel (PTX) and SP-LPS (nontoxic derivative of lipopolysaccharide) were selected as anticancer drug and immunostimulant respectively. Poly(lactic-co-glycolic acid) (PLGA) based PTX and SP-LPS containing nanoparticles (TLNP) were prepared by the double-emulsion method (w/o/w) and characterized in terms of size, zeta potential and transmission electron microscopy (TEM). The release behavior of PTX and SP-LPS from the TLNP exhibited a biphasic pattern characterized by an initial burst followed by slow continuous release. In vitro anticancer activity of TLNP was found to be higher compared to PTX when studied in a tumor cell-splenocyte coculture system. TLNP activated murine monocytes induced the secretion of various proinflammatory cytokines. After iv administration of TLNP in tumor bearing C57BL/6 mice, the amount of PTX in the tumor mass was found to be higher in TLNP treated mice as compared to commercial Taxol group at all time points studied. In vitro studies suggest that nanoparticles containing PTX and SP-LPS have both direct cytotoxicity and immunostimulatory activity. Hence this might have potential as a chemo-immunotherapeutic formulation against cancer with advantage over present day chemotherapy with Taxol, in terms of tumor targeting, less toxicity and immunostimulation.
Collapse
Affiliation(s)
- Aniruddha Roy
- Product Development Cell-I, National Institute of Immunology, New Delhi, 110067, India
| | | | | | | |
Collapse
|
20
|
Bellati F, Napoletano C, Gasparri ML, Panici PB, Nuti M. Immunologic systemic effect of neoadjuvant chemotherapy requires investigation before tumor-associated lymphocytes can be introduced in breast cancer treatment algorithm. J Clin Oncol 2010; 28:e471-2; author reply e473. [PMID: 20585098 DOI: 10.1200/jco.2010.27.9984] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
21
|
Pfannenstiel LW, Lam SSK, Emens LA, Jaffee EM, Armstrong TD. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol 2010; 263:79-87. [PMID: 20346445 DOI: 10.1016/j.cellimm.2010.03.001] [Citation(s) in RCA: 127] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 02/24/2010] [Accepted: 03/01/2010] [Indexed: 12/19/2022]
Abstract
Subclinical doses of Paclitaxel (PTX) given 1day prior to a HER-2/neu (neu)-targeted, granulocyte-macrophage colony stimulating factor (GM-CSF)-secreting whole-cell vaccine enhances neu-specific T cell responses and slows neu(+) tumor growth in tolerized HER-2/neu (neu-N) mice. We demonstrate that co-administration of PTX and Cyclophosphamide (CY) synergizes to slow tumor growth, and that in vitro, DC precursors exposed to PTX before LPS maturation results in greater co-stimulatory molecule expression, IL-12 production, and the ability to induce CD8(+) T cells with enhanced lytic activity against neu(+) tumors. PTX treatment also enhances maturation marker expression on CD11c(+) DCs isolated from vaccine-draining lymph nodes. Ex vivo, these DCs activate CD8(+) T cells with greater lytic capability than DC's from vaccine alone-treated neu-N mice. Finally, PTX treatment results in enhanced antigen-specific, IFN-gamma-secreting CD8(+) T cells in vivo. Thus, administration of PTX with a tumor vaccine improves T cell priming through enhanced maturation of DC.
Collapse
Affiliation(s)
- Lukas W Pfannenstiel
- Department of Oncology, Sidney Kimmel Cancer Center, Division of Immunology, Johns Hopkins University, USA
| | | | | | | | | |
Collapse
|
22
|
Javeed A, Ashraf M, Riaz A, Ghafoor A, Afzal S, Mukhtar MM. Paclitaxel and immune system. Eur J Pharm Sci 2009; 38:283-90. [DOI: 10.1016/j.ejps.2009.08.009] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 08/29/2009] [Indexed: 12/17/2022]
|
23
|
Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. Br J Cancer 2009; 101:1513-21. [PMID: 19861998 PMCID: PMC2778517 DOI: 10.1038/sj.bjc.6605274] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: The aim of this study was to investigate the prognostic effect of tumour-infiltrating lymphocytes (TILs) in serous stage III ovarian carcinoma to determine TIL clonality and to correlate this to Her2/neu expression. Methods: Formalin-fixed and paraffin-embedded ovarian carcinomas were examined for CD20-, CD3-, CD4- and CD8-positive lymphocytes (n=100), and for Her2/neu-positive tumour cells (n=55/100) by immunohistochemistry. Clonality analysis was carried out by T-cell receptor γ (TCRγ) gene rearrangements (n=93/100). Statistical analyses included experimental and clinico-pathological variables, as well as disease-free (DFS) and overall (OS) survival. Results: CD20-positive B lymphocytes were present in 57.7% (stromal)/33.0% (intraepithelial) and CD3-positive T lymphocytes in 99.0% (stromal)/90.2% (intraepithelial) of ovarian carcinomas. Intraepithelial CD3-positive T lymphocytes were correlated with improved DFS in optimally debulked patients (P=0.0402). Intraepithelial CD8-positive T lymphocytes were correlated with improved OS in all optimally debulked patients (P=0.0201) and in those undergoing paclitaxel/carboplatin therapy (P=0.0092). Finally, rarified and clonal TCRγ gene rearrangements were detected in 37 out of 93 (39.8%) and 15 out of 93 (16.1%) cases, respectively. This was marginally associated with improved DFS (P=0.0873). Despite a significant correlation of HER2/neu status and intraepithelial CD8-positive lymphocytes (P=0.0264), this was non-directional (R=−0.257; P=0.0626). Conclusion: Improved survival of ovarian cancer patients is related to the infiltration, clonal selection and intraepithelial persistence of T lymphocytes.
Collapse
|
24
|
Mine T, Matsueda S, Li Y, Tokumitsu H, Gao H, Danes C, Wong KK, Wang X, Ferrone S, Ioannides CG. Breast cancer cells expressing stem cell markers CD44+ CD24 lo are eliminated by Numb-1 peptide-activated T cells. Cancer Immunol Immunother 2009; 58:1185-94. [PMID: 19048252 PMCID: PMC2726795 DOI: 10.1007/s00262-008-0623-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 10/29/2008] [Indexed: 12/22/2022]
Abstract
Cancer stem cells (CSC) are resistant to chemo- and radiotherapy. To eliminate cells with phenotypic markers of CSC-like we characterized: (1) expression of CD44, CD24, CD133 and MIC-A/B (NKG2 receptors) in breast (MCF7) and ovarian (SK-OV-3) cells resistant to gemcitabine (GEM), paclitaxel (PTX) and 5-fluorouracil (5-FU) and (2) their elimination by Numb- and Notch-peptide activated CTL. The number of cells in all populations with the luminal CSC phenotype [epithelial specific antigen(+) (ESA) CD44(hi) CD24(lo), CD44(hi) CD133(+), and CD133(+) CD24(lo)] increased in drug-resistant MCF7 and SK-OV-3 cells. Similarly, the number of cells with expressed MIC-A/B increased 4 times in drug-resistant tumor cells compared with drug-sensitive cells. GEM(Res) MCF7 cells had lower levels of the Notch-1-extracellular domain (NECD) and Notch trans-membrane intracellular domain (TMIC) than GEM(Sens) MCF7. The levels of Numb, and Numb-L-[P]-Ser(265) were similar in GEM(Res) and GEM(Sens) MCF7 cells. Only the levels of Numb-L (long)-Ser(295) decreased slightly. This finding suggests that Notch-1 cleavage to TMIC is inhibited in GEM(Res) MCF7 cells. PBMC activated by natural immunogenic peptides Notch-1 (2112-2120) and Numb-1 (87-95) eliminated NICD(positive), CD24(hi) CD24(lo) MCF7 cells. It is likely that the immunogenic Numb-1 peptide in MCF7 cells originated from Numb, [P]-lated by an unknown kinase, because staurosporine but not wortmannin and MAPK-inhibitors decreased peptide presentation. Numb and Notch are antagonistic proteins which degrade each other to stop and activate cell proliferation, respectively. Their peptides are presented alternatively. Targeting both antagonistic proteins should be useful to prevent metastases in patients whose tumors are resistant to conventional treatments.
Collapse
MESH Headings
- Anticarcinogenic Agents/pharmacology
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- CD24 Antigen/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Female
- GPI-Linked Proteins
- Humans
- Hyaluronan Receptors/immunology
- Immunotherapy, Active
- Immunotherapy, Adoptive
- Intercellular Signaling Peptides and Proteins/immunology
- Intercellular Signaling Peptides and Proteins/metabolism
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Nerve Tissue Proteins/immunology
- Nerve Tissue Proteins/metabolism
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/metabolism
- Peptides/immunology
- Receptor, Notch1/immunology
- Receptor, Notch1/metabolism
Collapse
Affiliation(s)
- Takashi Mine
- Department of Gynecologic Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vicari AP, Luu R, Zhang N, Patel S, Makinen SR, Hanson DC, Weeratna RD, Krieg AM. Paclitaxel reduces regulatory T cell numbers and inhibitory function and enhances the anti-tumor effects of the TLR9 agonist PF-3512676 in the mouse. Cancer Immunol Immunother 2009; 58:615-28. [PMID: 18802696 PMCID: PMC11030133 DOI: 10.1007/s00262-008-0586-2] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2008] [Accepted: 08/26/2008] [Indexed: 12/20/2022]
Abstract
The anti-tumor properties of Toll-like receptor (TLR) 9 agonist CpG oligodeoxynucleotides (ODN) are enhanced by combinations with several cytotoxic chemotherapy regimens. The mechanisms of this added benefit, however, remain unclear. We now report that, similar to the depletion of regulatory T cells (Treg) using anti-CD25, paclitaxel increased the anti-tumor effect of the TLR9 agonist PF-3512676 in a CD8(+) T cell-dependent fashion. Paclitaxel treatment decreased Treg numbers in a TLR4-independent fashion, and preferentially affected cycling Treg expressing high levels of FoxP3. The paclitaxel-induced reduction in Treg FoxP3 expression was associated with reduced inhibitory function. Adoptively transferred tumor-antigen specific CD8(+) T cells proliferated better in mice treated with paclitaxel and their recruitment in the tumor was increased. However, the systemic frequency of PF-3512676-induced tumor-antigen specific effector CD8(+) T cells decreased with paclitaxel, suggesting opposite effects of paclitaxel on the anti-tumor response. Finally, gene expression profiling and studies of tumor-associated immune cells revealed a complex modulation of the PF-3512676-induced immune response by paclitaxel, including a decrease of IL-10 expression and an increase in IL-17-secreting CD4(+) T cells. Collectively, these data suggest that paclitaxel combined with PF-3512676 may not only promote a better anti-tumor CD8(+) response though increased recruitment in the tumor, possibly through Treg depletion and suppression, but also exerts more complex immune modulatory effects.
Collapse
Affiliation(s)
- Alain P Vicari
- Coley Pharmaceutical Group-A Pfizer Company, 340 Terry Fox Drive, suite 200, Ottawa, ON, K2K 3A2, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Ovarian cancer remains a challenging disease for which improved treatments are urgently needed. Most patients present with advanced disease that is highly responsive to surgery combined with platinum- and taxane-based chemotherapy, with a state of minimal residual disease being achieved in many cases. However, chemotherapy-resistant recurrent tumors typically appear within 1-5 years and are ultimately fatal. Recently, several groups have shown that ovarian tumors are often infiltrated by activated T cells at the time of diagnosis, and patients with dense infiltrates of CD3+CD8+ T cells experience unexpectedly favorable progression-free and overall survival. Other cell types in the immune infiltrate oppose anti-tumor immunity, including CD4+CD25+FoxP3+ regulatory T cells, CD8+ regulatory T cells, macrophages, and dendritic cells. The composition of immune infiltrates is shaped by the expression of cytokines, chemokines, antigens, major histocompatibility complex molecules, and costimulatory molecules. The relationship between these various immunological factors is reviewed here with a strong emphasis on outcomes data so as to create a knowledge base that is well grounded in clinical reality. With improved understanding of the functional properties of natural CD8+ T-cell responses to ovarian cancer, there is great potential to improve clinical outcomes by amplifying host immunity.
Collapse
Affiliation(s)
- Brad H Nelson
- Trev & Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, BC, Canada.
| |
Collapse
|
27
|
Abstract
The transcription factor glioma-associated antigen-1 (Gli-1) mediates activation of the sonic hedgehog (Shh) pathway, a process that precedes the transformation of tissue stem cells into cancerous stem cells and that is involved in early and late epithelial tumorigenesis. Hypothesizing that targeting the 3'-untranslated region (3'-UTR) of Gli-1 mRNA would effectively inhibit epithelial tumor cell proliferation, we evaluated several complementary miRNA molecules for their ability to do so. The synthetic miRNAs and corresponding duplex/small temporal RNAs were introduced as 3-nucleotide (nt) loops into GU-rich portions of the 3'UTR Gli-1 sequence. One particular miRNA (miRNA Gli-1-3548) and its corresponding duplex (Duplex 3548) significantly inhibited proliferation of Gli-1+ ovarian (SK-OV-3) and pancreatic (MiaPaCa-2) tumor cells by delaying cell division and activating late apoptosis in MiaPaCa-2 cells. Here, we describe the design of effective miRNA sequences and their applications as anti-gene agents.
Collapse
|
28
|
Tumor-infiltrating T cells correlate with NY-ESO-1-specific autoantibodies in ovarian cancer. PLoS One 2008; 3:e3409. [PMID: 18923710 PMCID: PMC2561074 DOI: 10.1371/journal.pone.0003409] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2008] [Accepted: 09/17/2008] [Indexed: 12/20/2022] Open
Abstract
Background Tumor-infiltrating CD8+ T cells are correlated with prolonged progression-free and overall survival in epithelial ovarian cancer (EOC). A significant fraction of EOC patients mount autoantibody responses to various tumor antigens, however the relationship between autoantibodies and tumor-infiltrating T cells has not been investigated in EOC or any other human cancer. We hypothesized that autoantibody and T cell responses may be correlated in EOC and directed toward the same antigens. Methodology and Principal Findings We obtained matched serum and tumor tissue from 35 patients with high-grade serous ovarian cancer. Serum samples were assessed by ELISA for autoantibodies to the common tumor antigen NY-ESO-1. Tumor tissue was examined by immunohistochemistry for expression of NY-ESO-1, various T cell markers (CD3, CD4, CD8, CD25, FoxP3, TIA-1 and Granzyme B) and other immunological markers (CD20, MHC class I and MHC class II). Lymphocytic infiltrates varied widely among tumors and included cells positive for CD3, CD8, TIA-1, CD25, FoxP3 and CD4. Twenty-six percent (9/35) of patients demonstrated serum IgG autoantibodies to NY-ESO-1, which were positively correlated with expression of NY-ESO-1 antigen by tumor cells (r = 0.57, p = 0.0004). Autoantibodies to NY-ESO-1 were associated with increased tumor-infiltrating CD8+, CD4+ and FoxP3+ cells. In an individual HLA-A2+ patient with autoantibodies to NY-ESO-1, CD8+ T cells isolated from solid tumor and ascites were reactive to NY-ESO-1 by IFN-γ ELISPOT and MHC class I pentamer staining. Conclusion and Significance We demonstrate that tumor-specific autoantibodies and tumor-infiltrating T cells are correlated in human cancer and can be directed against the same target antigen. This implies that autoantibodies may collaborate with tumor-infiltrating T cells to influence clinical outcomes in EOC. Furthermore, serological screening methods may prove useful for identifying clinically relevant T cell antigens for immunotherapy.
Collapse
|
29
|
Chu CS, Kim SH, June CH, Coukos G. Immunotherapy opportunities in ovarian cancer. Expert Rev Anticancer Ther 2008; 8:243-57. [PMID: 18279065 DOI: 10.1586/14737140.8.2.243] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is responsible for the majority of gynecologic cancer deaths and despite the highest standard of multimodality therapy with surgery and cytotoxic chemotherapy, long-term survival remains low. With compelling evidence that epithelial ovarian cancer is an immunogenic tumor capable of stimulating an antitumor immune response, renewed efforts to develop immune therapies to augment the efficacy of traditional therapies are underway. Current immunotherapies focus on varied modes of antitumor vaccine development, particularly with the use of dendritic cell vaccines, effective methods for adoptive T-cell transfer and combinatorial approaches with immune modulatory therapy subverting natural tolerance mechanisms or boosting effector mechanisms. Additional combinatorial approaches include the use of cytokines and/or chemotherapy with immune therapy.
Collapse
Affiliation(s)
- Christina S Chu
- University of Pennsylvania, Division of Gynecologic Oncology, Center for Research on Ovarian Cancer, PA, USA.
| | | | | | | |
Collapse
|
30
|
López-Larrea C, Suárez-Alvarez B, López-Soto A, López-Vázquez A, Gonzalez S. The NKG2D receptor: sensing stressed cells. Trends Mol Med 2008; 14:179-89. [PMID: 18353724 DOI: 10.1016/j.molmed.2008.02.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2007] [Revised: 02/13/2008] [Accepted: 02/13/2008] [Indexed: 12/22/2022]
Abstract
The activating killer cell lectin-like receptor NKG2D plays a key role in the natural killer (NK) cell-mediated lysis of tumours and infected cells. Unlike other receptors, the ligands recognised by NKG2D are 'induced-self' ligands on stressed cells. This system requires precise regulation because inappropriate expression of NKG2D ligands might compromise NK cell activation. For therapeutic purposes it is essential to understand the mechanisms that regulate the expression and function of the NKG2D system. This review focuses on the importance of the signalling pathways involved in the regulation of the NKG2D receptor and its ligand expression in arming the immune response against infected or tumour cells and for the identification of new molecular targets and therapeutic strategies.
Collapse
Affiliation(s)
- Carlos López-Larrea
- Department of Immunology, Histocompatibility Unit, Hospital Universitario Central de Asturias, Julian Claveria Street, 33006 Oviedo, Spain.
| | | | | | | | | |
Collapse
|