101
|
Apetoh L, Ladoire S, Coukos G, Ghiringhelli F. Combining immunotherapy and anticancer agents: the right path to achieve cancer cure? Ann Oncol 2015; 26:1813-1823. [PMID: 25922066 DOI: 10.1093/annonc/mdv209] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/17/2015] [Indexed: 12/31/2022] Open
Abstract
Recent clinical trials revealed the impressive efficacy of immunological checkpoint blockade in different types of metastatic cancers. Such data underscore that immunotherapy is one of the most promising strategies for cancer treatment. In addition, preclinical studies provide evidence that some cytotoxic drugs have the ability to stimulate the immune system, resulting in anti-tumor immune responses that contribute to clinical efficacy of these agents. These observations raise the hypothesis that the next step for cancer treatment is the combination of cytotoxic agents and immunotherapies. The present review aims to summarize the immune-mediated effects of chemotherapeutic agents and their clinical relevance, the biological and clinical features of immune checkpoint blockers and finally, the preclinical and clinical rationale for novel therapeutic strategies combining anticancer agents and immune checkpoint blockers.
Collapse
Affiliation(s)
- L Apetoh
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France
| | - S Ladoire
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France
| | - G Coukos
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - F Ghiringhelli
- Lipids, Nutrition, Cancer, INSERM, U866, Dijon; Department of Medicine, Université de Bourgogne, Dijon; Department of Oncology, Centre Georges François Leclerc, Dijon, France.
| |
Collapse
|
102
|
Zheng Y, Dou Y, Duan L, Cong C, Gao A, Lai Q, Sun Y. Using chemo-drugs or irradiation to break immune tolerance and facilitate immunotherapy in solid cancer. Cell Immunol 2015; 294:54-9. [PMID: 25687508 DOI: 10.1016/j.cellimm.2015.02.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/01/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
The immunity is dual host-protective and tumor-promoting in cancer development and progression. Many immune suppressive cells and cytokines in microenvironment can prevent cytotoxic T lymphocytes (CTL) and natural killer cells (NK) from killing tumor cells. Chemotherapy drugs and irradiation have been reported helpful in breaking immune tolerance and creating microenvironment for adoptive cell therapy. Low-dose cyclophosphamide or gemcitabine therapy can selectively deplete T regulatory cells (Treg). Paclitaxel can alter cytokine network at the tumor site, and 5-fluorouracil shows a pronounced effect on myeloid-derived suppressor cells (MDSC) depletion. Local tumor irradiation and total body irradiation (TBI) can also affect tumor microenvironment and facilitate immunotherapy. In this review, we summarize the particular effects of these agents and methods on immunomodulation, as well as their potential values in immunotherapy. The combination with immunotherapy represents a novel therapeutic strategy.
Collapse
Affiliation(s)
- Yawen Zheng
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yan Dou
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Lili Duan
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Changsheng Cong
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Qinghua Lai
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yuping Sun
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, China.
| |
Collapse
|
103
|
Miolo G, Muraro E, Martorelli D, Lombardi D, Scalone S, Spazzapan S, Massarut S, Perin T, Viel E, Comaro E, Talamini R, Bidoli E, Turchet E, Crivellari D, Dolcetti R. Anthracycline-free neoadjuvant therapy induces pathological complete responses by exploiting immune proficiency in HER2+ breast cancer patients. BMC Cancer 2014; 14:954. [PMID: 25512030 PMCID: PMC4302069 DOI: 10.1186/1471-2407-14-954] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 12/11/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Neoadjuvant Chemotherapy (NC) including trastuzumab induces a high rate of pathological Complete Responses (pCR) in patients with locally advanced HER2-overexpressing Breast Cancer (BC), but is penalized by a severe cardiotoxicity when combined with anthracyclines. A phase II study was designed to assess whether an anthracycline-free NC regimen based on the early addition of trastuzumab to paclitaxel may increase the pCR rate without inducing severe cardiotoxicity in patients with locally advanced HER2-overexpressing BC. Immunomonitoring was performed to assess the contribution of patients' immunological background to the induction of clinical responses. METHODS Stage II-III HER2-positive BC patients received 24 weeks paclitaxel and trastuzumab NC, followed by 1 year adjuvant trastuzumab ± hormonal and/or radio-therapy. Assessment of pCR rate was the primary endpoint. A group of HER2-negative BC patients treated with neoadjuvant taxanes and anthracyclines was included. Serum levels of 10 cytokines and the efficiency of trastuzumab-mediated antibody-dependent cell cytotoxicity (ADCC) were monitored in vitro every 3 months. RESULTS From July 2006 to February 2013, we enrolled 109 patients including 46 evaluable HER2-positive cases. A pCR rate of 50% was reached and no severe cardiotoxicity occurred. Serum cytokine profiling revealed only an IL-10 decrease (P = 0.02) in patients achieving a partial response, while HER2-negative patients disclosed marked cytokines changes. Compared to the unfavourable F/F genotype, patients carrying the V allele in the FcγRIIIa-158 polymorphism showed a higher efficacy of trastuzumab-ADCC throughout treatment (P ≤0.05). CONCLUSIONS In the absence of anthracyclines, trastuzumab and paclitaxel induced a high rate of pCR, exploiting the synergy between the immunomodulating properties of these drugs and the retained immunological proficiency of patients with HER2-overexpressing BC. TRIAL REGISTRATION Trial registration number: NCT02307227, registered on ClinicalTrials.gov (http://www.clinicaltrials.gov, November 26, 2014).
Collapse
Affiliation(s)
- Gianmaria Miolo
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elena Muraro
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Debora Martorelli
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Davide Lombardi
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Simona Scalone
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Simon Spazzapan
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Samuele Massarut
- />Division of Breast Surgical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Tiziana Perin
- />Pathology Unit, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elda Viel
- />Cardiology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elisa Comaro
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Renato Talamini
- />Unit of Epidemiology and Biostatistics, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Ettore Bidoli
- />Unit of Epidemiology and Biostatistics, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Elisa Turchet
- />Scientific Direction, CRO Aviano, IRCCS, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Diana Crivellari
- />Department of Medical Oncology, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| | - Riccardo Dolcetti
- />Cancer Bio-Immunotherapy Unit, Department of Medical Oncology, C.R.O, National Cancer Institute, Via F. Gallini 2, 33081 Aviano, PN Italy
| |
Collapse
|
104
|
Huang ZZ, Li D, Liu CC, Cui Y, Zhu HQ, Zhang WW, Li YY, Xin WJ. CX3CL1-mediated macrophage activation contributed to paclitaxel-induced DRG neuronal apoptosis and painful peripheral neuropathy. Brain Behav Immun 2014; 40:155-65. [PMID: 24681252 DOI: 10.1016/j.bbi.2014.03.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/17/2014] [Accepted: 03/20/2014] [Indexed: 02/06/2023] Open
Abstract
Painful peripheral neuropathy is a dose-limiting side effect of paclitaxel therapy, which hampers the optimal clinical management of chemotherapy in cancer patients. Currently the underlying mechanisms remain largely unknown. Here we showed that the clinically relevant dose of paclitaxel (3×8mg/kg, cumulative dose 24mg/kg) induced significant upregulation of the chemokine CX3CL1 in the A-fiber primary sensory neurons in vivo and in vitro and infiltration of macrophages into the dorsal root ganglion (DRG) in rats. Paclitaxel treatment also increased cleaved caspase-3 expression, induced the loss of primary afferent terminal fibers and decreased sciatic-evoked A-fiber responses in the spinal dorsal horn, indicating DRG neuronal apoptosis induced by paclitaxel. In addition, the paclitaxel-induced DRG neuronal apoptosis occurred exclusively in the presence of macrophage in vitro study. Intrathecal or systemic injection of CX3CL1 neutralizing antibody blocked paclitaxel-induced macrophage recruitment and neuronal apoptosis in the DRG, and also attenuated paclitaxel-induced allodynia. Furthermore, depletion of macrophage by systemic administration of clodronate inhibited paclitaxel-induced allodynia. Blocking CX3CL1 decreased activation of p38 MAPK in the macrophage, and inhibition of p38 MAPK activity blocked the neuronal apoptosis and development of mechanical allodynia induced by paclitaxel. These findings provide novel evidence that CX3CL1-recruited macrophage contributed to paclitaxel-induced DRG neuronal apoptosis and painful peripheral neuropathy.
Collapse
Affiliation(s)
- Zhen-Zhen Huang
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Dai Li
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Cui-Cui Liu
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Yu Cui
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - He-Quan Zhu
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Wen-Wen Zhang
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Yong-Yong Li
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China
| | - Wen-Jun Xin
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd. 2, 510080, China.
| |
Collapse
|
105
|
Gutbrodt KL, Casi G, Neri D. Antibody-based delivery of IL2 and cytotoxics eradicates tumors in immunocompetent mice. Mol Cancer Ther 2014; 13:1772-6. [PMID: 24759429 DOI: 10.1158/1535-7163.mct-14-0105] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates are increasingly being used for cancer therapy, but little is known about their ability to promote anticancer immunity, which may lead to long-lasting remissions. We investigated the therapeutic effect of antibody-based pharmacodelivery of cemadotin, a cytotoxic drug, and IL2, a strong proinflammatory cytokine. Using the F8 antibody, which selectively localizes to the tumor neovasculature, combination treatment led to tumor eradication, in a process dependent on CD8(+) T cells and natural killer cells in the C1498 syngeneic mouse model of acute myelogenous leukemia. The clinical combination of antibody-drug conjugates and antibody-cytokine proteins should be facilitated by their orthogonal toxicity profiles.
Collapse
Affiliation(s)
- Katrin L Gutbrodt
- Authors' Affiliations: Department of Chemistry and Applied Biosciences, ETH Zürich, Zurich; and
| | | | - Dario Neri
- Authors' Affiliations: Department of Chemistry and Applied Biosciences, ETH Zürich, Zurich; and
| |
Collapse
|
106
|
Zeng S, Wu F, Li B, Song X, Zheng Y, He G, Peng C, Huang W. Synthesis, characterization, and evaluation of a novel amphiphilic polymer RGD-PEG-Chol for target drug delivery system. ScientificWorldJournal 2014; 2014:546176. [PMID: 24578646 PMCID: PMC3918714 DOI: 10.1155/2014/546176] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/24/2013] [Indexed: 02/05/2023] Open
Abstract
An amphiphilic polymer RGD-PEG-Chol which can be produced in large scale at a very low cost has been synthesized successfully. The synthesized intermediates and final products were characterized and confirmed by ¹H nuclear magnetic resonance spectrum (¹H NMR) and Fourier transform infrared spectrum (FT-IR). The paclitaxel- (PTX-) loaded liposomes based on RGD-PEG-Chol were then prepared by film formation method. The liposomes had a size within 100 nm and significantly enhanced the cytotoxicity of paclitaxel to B16F10 cell as demonstrated by MTT test (IC₅₀ = 0.079 μg/mL of RGD-modified PTX-loaded liposomes compared to 9.57 μg/mL of free PTX). Flow cytometry analysis revealed that the cellular uptake of coumarin encapsulated in the RGD-PEG-Chol modified liposome was increased for HUVEC cells. This work provides a reasonable, facile, and economic approach to prepare peptide-modified liposome materials with controllable performances and the obtained linear RGD-modified PTX-loaded liposomes might be attractive as a drug delivery system.
Collapse
Affiliation(s)
- Shi Zeng
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Fengbo Wu
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Bo Li
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Xiangrong Song
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Yu Zheng
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Gu He
- State Key Laboratory of Biotherapy and Department of Pharmacy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Cheng Peng
- State Key Laboratory Breeding Base of Systematic research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Huang
- State Key Laboratory Breeding Base of Systematic research, Development and Utilization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
107
|
Bracci L, Schiavoni G, Sistigu A, Belardelli F. Immune-based mechanisms of cytotoxic chemotherapy: implications for the design of novel and rationale-based combined treatments against cancer. Cell Death Differ 2014; 21:15-25. [PMID: 23787994 PMCID: PMC3857622 DOI: 10.1038/cdd.2013.67] [Citation(s) in RCA: 673] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/07/2013] [Accepted: 05/14/2013] [Indexed: 02/06/2023] Open
Abstract
Conventional anticancer chemotherapy has been historically thought to act through direct killing of tumor cells. This concept stems from the fact that cytotoxic drugs interfere with DNA synthesis and replication. Accumulating evidence, however, indicates that the antitumor activities of chemotherapy also rely on several off-target effects, especially directed to the host immune system, that cooperate for successful tumor eradication. Chemotherapeutic agents stimulate both the innate and adaptive arms of the immune system through several modalities: (i) by promoting specific rearrangements on dying tumor cells, which render them visible to the immune system; (ii) by influencing the homeostasis of the hematopoietic compartment through transient lymphodepletion followed by rebound replenishment of immune cell pools; (iii) by subverting tumor-induced immunosuppressive mechanisms and (iv) by exerting direct or indirect stimulatory effects on immune effectors. Among the indirect ways of immune cell stimulation, some cytotoxic drugs have been shown to induce an immunogenic type of cell death in tumor cells, resulting in the emission of specific signals that trigger phagocytosis of cell debris and promote the maturation of dendritic cells, ultimately resulting in the induction of potent antitumor responses. Here, we provide an extensive overview of the multiple immune-based mechanisms exploited by the most commonly employed cytotoxic drugs, with the final aim of identifying prerequisites for optimal combination with immunotherapy strategies for the development of more effective treatments against cancer.
Collapse
Affiliation(s)
- L Bracci
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - G Schiavoni
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - A Sistigu
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - F Belardelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
108
|
Ogden A, Rida PCG, Reid MD, Aneja R. Interphase microtubules: chief casualties in the war on cancer? Drug Discov Today 2013; 19:824-9. [PMID: 24201225 DOI: 10.1016/j.drudis.2013.10.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/09/2013] [Accepted: 10/28/2013] [Indexed: 12/13/2022]
Abstract
Microtubule-targeting agents (MTAs) profoundly affect interphase cells, such as by disrupting axonal transport, transcription, translation, mitochondrial permeability, immune cell function, directional migration and centrosome clustering. This finding is antithetical to the conventionally held notion that MTAs act on mitosis to trigger arrest-mediated apoptotic cell death. Furthermore, the paucity of mitotic cells in patient tumors and lack of correlation of MTA efficacy with tumor proliferation rate provide strong impetus to re-examine the mechanistic basis of action of MTAs, with an eye toward interphase activities. Whereas targeted antimitotics have unequivocally failed their promise across clinical studies, MTAs constitute a mainstay of chemotherapy. This paradox necessitates the conclusion that MTAs exert mitosis-independent effects, spurring a dramatic paradigm shift in our understanding of the mode of action of MTAs.
Collapse
Affiliation(s)
- Angela Ogden
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | - Michelle D Reid
- Department of Pathology, Emory University Hospital, Atlanta, GA 30322, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
109
|
Seo AN, Lee HJ, Kim EJ, Kim HJ, Jang MH, Lee HE, Kim YJ, Kim JH, Park SY. Tumour-infiltrating CD8+ lymphocytes as an independent predictive factor for pathological complete response to primary systemic therapy in breast cancer. Br J Cancer 2013; 109:2705-13. [PMID: 24129232 PMCID: PMC3833219 DOI: 10.1038/bjc.2013.634] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 09/22/2013] [Accepted: 09/24/2013] [Indexed: 12/31/2022] Open
Abstract
Background: Tumour-infiltrating lymphocytes (TILs) are known to be associated with response to primary systemic therapy (PST) in breast cancer. This study was conducted to assess the association of TIL subsets with pathological complete response (pCR) after PST in breast cancer in relation to breast cancer subtype, breast cancer stem cell (BCSC) phenotype and epithelial–mesenchymal transition (EMT). Methods: The pre-chemotherapeutic biopsy specimens of 153 breast cancer patients who underwent surgical resection after anthracycline- or anthracycline/taxane-based PST were analysed. TIL subsets (CD4+, CD8+, and FOXP3+ TILs), BCSC phenotype, and the expression of EMT markers were evaluated by immunohistochemistry and were correlated with pCR after PST. Results: Infiltration of CD4+ and CD8+ T lymphocytes was closely correlated with BCSC phenotype and EMT. High levels of CD4+, CD8+, and FOXP3+ TILs were associated with pCR, and CD8+ TILs were found to be an independent predictive factor for pCR. In addition, CD8+ TILs were associated with pCR irrespective of breast cancer subtype, CD44+/CD24− phenotype, EMT, and chemotherapeutic regimen in subgroup analyses. Conclusion: These findings indicate that CD8+ cytotoxic T lymphocytes are a key component of TILs associated with chemo-response and can be used as a reliable predictor of response to anthracycline- or anthracycline/taxane-based PST in breast cancer.
Collapse
Affiliation(s)
- A N Seo
- 1] Department of Pathology, Seoul National University Bundang Hospital, 300 Gumi-dong, Bundang-gu, Seongnam-si, Gyeonggi 463-707, Korea [2] Department of Pathology, Kyungpook National University School of Medicine, 680 Gukchaebosang-ro, Jung-gu, Daegu 700-842, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Rovida A, Castiglioni V, Decio A, Scarlato V, Scanziani E, Giavazzi R, Cesca M. Chemotherapy Counteracts Metastatic Dissemination Induced by Antiangiogenic Treatment in Mice. Mol Cancer Ther 2013; 12:2237-47. [DOI: 10.1158/1535-7163.mct-13-0244] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
111
|
Abstract
Nanoscale objects, whether of biologic origin or synthetically created, are being developed into devices for a variety of bionanotechnology diagnostic and pharmaceutical applications. However, the potential immunotoxicity of these nanomaterials and mechanisms by which they may induce adverse reactions have not received sufficient attention. Nanomaterials, depending on their characteristics and compositions, can interact with the immune system in several ways and either enhance or suppress immune system function. Cytokines perform pleiotropic functions to mediate and regulate the immune response and are generally recognized as biomarkers of immunotoxicity. While the specificity and validity of certain cytokines as markers of adverse immune response has been established for chemicals, small and macromolecular drugs, research on their applicability for predicting and monitoring the immunotoxicity of engineered nanomaterials is still ongoing. The goal of this review is to provide guidelines as to important cytokines that can be utilized for evaluating the immunotoxicity of nanomaterials and to highlight the role of those cytokines in mediating adverse reactions, which is of particular importance for the clinical development of nanopharmaceuticals and other nanotechnology-based products. Importantly, the rational design of nanomaterials of low immunotoxicity will be discussed, focusing on synthetic nanodevices, with emphasis on both the nanoparticle-forming materials and the embedded cargoes.
Collapse
Affiliation(s)
- Mahmoud Elsabahy
- Department of Chemistry, Department of Chemical Engineering, and Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut International Center of Nanomedicine, Al-Rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Karen L. Wooley
- Department of Chemistry, Department of Chemical Engineering, and Laboratory for Synthetic-Biologic Interactions, Texas A&M University, P.O. Box 30012, 3255 TAMU, College Station, Texas 77842-3012, United States
| |
Collapse
|
112
|
Janssen A, Beerling E, Medema R, van Rheenen J. Intravital FRET imaging of tumor cell viability and mitosis during chemotherapy. PLoS One 2013; 8:e64029. [PMID: 23691140 PMCID: PMC3654962 DOI: 10.1371/journal.pone.0064029] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 04/10/2013] [Indexed: 11/18/2022] Open
Abstract
Taxanes, such as docetaxel, are microtubule-targeting chemotherapeutics that have been successfully used in the treatment of cancer. Based on data obtained from cell cultures, it is believed that taxanes induce tumor cell death by specifically perturbing mitotic progression. Here, we report on data that suggest that this generally accepted view may be too simplified. We describe a high-resolution intravital imaging method to simultaneously visualize mitotic progression and the onset of apoptosis. To directly compare in vitro and in vivo data, we have visualized the effect of docetaxel on mitotic progression in mouse and human colorectal tumor cell lines both in vitro and in isogenic tumors in mice. We show that docetaxel-induced apoptosis in vitro occurs via mitotic cell death, whereas the vast majority of tumor cells in their natural environment die independent of mitotic defects. This demonstrates that docetaxel exerts its anti-tumor effects in vivo through means other than mitotic perturbation. The differences between in vitro and in vivo mechanisms of action of chemotherapeutics may explain the limited response to many of the anti-mitotic agents that are currently validated in clinical trials. Our data illustrate the requirement and power of our intravital imaging technique to study and validate the mode of action of chemotherapeutic agents in vivo, which will be essential to understand and improve their clinical efficacy.
Collapse
Affiliation(s)
- Aniek Janssen
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology and Cancer Genomics University Medical Center Utrecht, Utrecht, The Netherlands
| | - Evelyne Beerling
- Cancer Genomics, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - René Medema
- Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology and Cancer Genomics University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail: (JvR); (RM)
| | - Jacco van Rheenen
- Cancer Genomics, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
- * E-mail: (JvR); (RM)
| |
Collapse
|
113
|
De Palma M, Lewis CE. Macrophage regulation of tumor responses to anticancer therapies. Cancer Cell 2013; 23:277-86. [PMID: 23518347 DOI: 10.1016/j.ccr.2013.02.013] [Citation(s) in RCA: 820] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/21/2013] [Accepted: 02/15/2013] [Indexed: 10/27/2022]
Abstract
Tumor-associated macrophages (TAMs) promote key processes in tumor progression, like angiogenesis, immunosuppression, invasion, and metastasis. Increasing studies have also shown that TAMs can either enhance or antagonize the antitumor efficacy of cytotoxic chemotherapy, cancer-cell targeting antibodies, and immunotherapeutic agents--depending on the type of treatment and tumor model. TAMs also drive reparative mechanisms in tumors after radiotherapy or treatment with vascular-targeting agents. Here, we discuss the biological significance and clinical implications of these findings, with an emphasis on novel approaches that effectively target TAMs to increase the efficacy of such therapies.
Collapse
Affiliation(s)
- Michele De Palma
- The Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| | | |
Collapse
|
114
|
Ramakrishnan R, Gabrilovich DI. Novel mechanism of synergistic effects of conventional chemotherapy and immune therapy of cancer. Cancer Immunol Immunother 2013; 62:405-10. [PMID: 23423351 PMCID: PMC11029489 DOI: 10.1007/s00262-012-1390-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/24/2012] [Indexed: 12/12/2022]
Abstract
There is mounting evidence to support the use of a combination of immunotherapy with chemotherapy in the treatment of various types of cancers. However, the mechanism(s), by which these modalities are synergized, are not fully understood. In this review, we discuss several possible mechanisms of the combined effect of immunotherapy and chemotherapy of cancer. We will examine various aspects of this issue such as the combination of different treatment options, the dosage for each arm of treatment, and, more importantly, the timing and sequence of the administration of these treatments.
Collapse
Affiliation(s)
- Rupal Ramakrishnan
- H. Lee Moffitt Cancer Center and Research Institute, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612 USA
| | - Dmitry I. Gabrilovich
- H. Lee Moffitt Cancer Center and Research Institute, MRC 2067, 12902 Magnolia Dr., Tampa, FL 33612 USA
| |
Collapse
|
115
|
Peng RQ, Ding Y, Zhang X, Liao Y, Zheng LM, Zhang XS. A pilot study of paclitaxel combined with gemcitabine followed by interleukin-2 and granulocyte macrophage colony-stimulating factor for patients with metastatic melanoma. Cancer Biol Ther 2012; 13:1443-8. [PMID: 22954698 DOI: 10.4161/cbt.22005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
It has been suggested that paclitaxel and gemcitabine modulate the immune system. This paper reports the safety and efficacy of paclitaxel plus gemcitabine followed by interleukin-2 (IL-2)and granulocyte macrophage colony-stimulating factor (GM-CSF), the PGIG chemobiotherapy, for patients with metastatic melanoma. All patients received 175 mg/m ( 2) paclitaxel on day 1 and 800 mg/m ( 2) gemcitabine on day two. IL-2 and GM-CSF were administered from day 4 to day 8 at a dosage of 2 MIU/m ( 2) and 100 μg, respectively. The PGIG chemobiotherapy was repeated every 21 d. Serum cytokine levels at baseline and at the end of the second cycle were measured via flow cytometry. Twenty-seven patients with metastatic melanoma accepted PGIG chemobiotherapy from August 2009 to March 2011. There were five patients that exhibited a partial response, 14 patients that exhibited a stable response and eight that displayed progressive disease. Therefore, the response rate was 18.5%, and the disease control rate was 70.4%. The median time to progression and median survival were 4 mo and 8 mo, respectively. The one-year and two-year survival rates were 25.9% and 18.5%, respectively. Frequent side effects included chills, fever, arthralgia, rash and pruritus. Among the 13 patients who experienced a rash and pruritus and the 14 patients who did not suffer from this side effect, the response rates and disease control rates were 30.8% vs 7.1% and 77% vs 64.2%, respectively. No relationship between serum IL-6 levels, clinical response, and either skin side effect was observed. The PGIG chemobiotherapy is safe and effective for the treatment of patients with advanced melanoma, but randomized trials are necessary to validate this effect.
Collapse
Affiliation(s)
- Rui-Qing Peng
- State Key Laboratory of Oncology in South China; Sun Yat-sen University; Guangzhou, China
| | | | | | | | | | | |
Collapse
|
116
|
Slovin SR. Toward maximizing immunotherapy in metastatic castration-resistant prostate cancer - rationale for combinatorial approaches using chemotherapy. Front Oncol 2012; 2:43. [PMID: 22662316 PMCID: PMC3362835 DOI: 10.3389/fonc.2012.00043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 04/12/2012] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer is particularly suited for active immunotherapy because of the expression of a distinctive number of antigens which are overexpressed on prostate cancer cells and cell lines. There is evidence in this disease that tumors promote immune tolerance starting early in the disease course. As such, chemotherapy, by suppressing tumors and activating immune system homeostatic mechanisms, may help overcome this tumor-induced immune tolerance. Sipuleucel-T which has recently been approved in the US, is an autologous cellular product immunotherapy that induces immune activity likely through activation of dendritic cells. This was associated with a survival benefit in the absence of significant toxicity. However, a post hoc analysis of phase III trial participants found a substantial survival benefit to receiving docetaxel some months after sipuleucel-T. However, another phase III immunotherapy trial combining a prostate cancer therapeutic vaccine GVAX plus docetaxel versus standard docetaxel therapy in advanced prostate cancer, observed a lower overall survival with the vaccine regimen. These trials highlight major unresolved questions concerning the optimum choice, dosing, and timing of chemotherapy relative to active immunotherapy and the overall merits of considering this approach. The ideal treatment approach remains unclear; advances in biomarker validation and trial design may likely improve our ability to assess biologic benefit irrespective of the development of true antitumor immunity.
Collapse
Affiliation(s)
- Susan R Slovin
- Genitourinary Oncology Service, Sidney Kimmel Center for Prostate and Urologic Cancers, Memorial Sloan-Kettering Cancer Center New York, NY, USA
| |
Collapse
|
117
|
Abstract
Cytotoxic cancer chemotherapy drugs are believed to gain selectivity by targeting cells that proliferate rapidly. However, the proliferation rate is low in many chemosensitive human cancers, and it is not clear how a drug that only kills dividing cells could promote tumor regression. Four potential solutions to this “proliferation rate paradox” are discussed for the microtubule-stabilizing drug paclitaxel: drug retention in tumors, killing of quiescent cells, targeting of noncancer cells in the tumor, and bystander effects. Testing these potential mechanisms of drug action will facilitate rational improvement of antimitotic chemotherapy and perhaps cytotoxic chemotherapy more generally.
Collapse
Affiliation(s)
- Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
118
|
Moschetta M, Pretto F, Berndt A, Galler K, Richter P, Bassi A, Oliva P, Micotti E, Valbusa G, Schwager K, Kaspar M, Trachsel E, Kosmehl H, Bani MR, Neri D, Giavazzi R. Paclitaxel Enhances Therapeutic Efficacy of the F8-IL2 Immunocytokine to EDA-Fibronectin–Positive Metastatic Human Melanoma Xenografts. Cancer Res 2012; 72:1814-24. [DOI: 10.1158/0008-5472.can-11-1919] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
119
|
Liu X, Zhao M, Wu K, Chai X, Yu H, Tao Z, Wang J. Immunomodulatory and anticancer activities of phenolics from emblica fruit (Phyllanthus emblica L.). Food Chem 2012. [DOI: 10.1016/j.foodchem.2011.09.063] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
120
|
Ye VM, Bhatia SK. Metabolic engineering for the production of clinically important molecules: Omega-3 fatty acids, artemisinin, and taxol. Biotechnol J 2011; 7:20-33. [PMID: 22021189 DOI: 10.1002/biot.201100289] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 08/28/2011] [Accepted: 09/02/2011] [Indexed: 11/07/2022]
Abstract
Driven by requirements for sustainability as well as affordability and efficiency, metabolic engineering of plants and microorganisms is increasingly being pursued to produce compounds for clinical applications. This review discusses three such examples of the clinical relevance of metabolic engineering: the production of omega-3 fatty acids for the prevention of cardiovascular disease; the biosynthesis of artemisinic acid, an anti-malarial drug precursor, for the treatment of malaria; and the production of the complex natural molecule taxol, an anti-cancer agent. In terms of omega-3 fatty acids, bioengineering of fatty acid metabolism by expressing desaturases and elongases, both in soybeans and oleaginous yeast, has resulted in commercial-scale production of these beneficial molecules. Equal success has been achieved with the biosynthesis of artemisinic acid at low cost for developing countries. This is accomplished through channeling the flux of the isoprenoid pathway to the specific genes involved in artemisinin biosynthesis. Efficient coupling of the isoprenoid pathway also leads to the construction of an Escherichia coli strain that produces a high titer of taxadiene-the first committed intermediate for taxol biosynthesis. These examples of synthetic biology demonstrate the versatility of metabolic engineering to bring new solutions to our health needs.
Collapse
Affiliation(s)
- Victor M Ye
- Health Promotion and Disease Prevention, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
121
|
Immune modulation by chemotherapy or immunotherapy to enhance cancer vaccines. Cancers (Basel) 2011; 3:3114-42. [PMID: 24212948 PMCID: PMC3759189 DOI: 10.3390/cancers3033114] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy has been a mainstay in cancer treatment for many years. Despite some success, the cure rate with chemotherapy remains unsatisfactory in some types of cancers, and severe side effects from these treatments are a concern. Recently, understanding of the dynamic interplay between the tumor and immune system has led to the development of novel immunotherapies, including cancer vaccines. Cancer vaccines have many advantageous features, but their use has been hampered by poor immunogenicity. Many developments have increased their potency in pre-clinical models, but cancer vaccines continue to have a poor clinical track record. In part, this could be due to an inability to effectively overcome tumor-induced immune suppression. It had been generally assumed that immune-stimulatory cancer vaccines could not be used in combination with immunosuppressive chemotherapies, but recent evidence has challenged this dogma. Chemotherapies could be used to condition the immune system and tumor to create an environment where cancer vaccines have a better chance of success. Other types of immunotherapies could also be used to modulate the immune system. This review will discuss how immune modulation by chemotherapy or immunotherapy could be used to bolster the effects of cancer vaccines and discuss the advantages and disadvantages of these treatments.
Collapse
|
122
|
André N, Padovani L, Pasquier E. Metronomic scheduling of anticancer treatment: the next generation of multitarget therapy? Future Oncol 2011; 7:385-94. [DOI: 10.2217/fon.11.11] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Metronomic scheduling of anticancer treatment (MSAT) is progressively gaining interest after the antiangiogenic properties of metronomic chemotherapy and its potential to overcome drug resistance was initially described in 2000. MSAT has now grown beyond the anticipated scope of antiangiogenic chemotherapy, with accumulating evidence demonstrating that these treatments may also act by stimulating an antitumor immune response and could ultimately lead to reinduction of tumor dormancy. An increasing number of drugs, not initially developed as anticancer agents, are currently being used in metronomic protocols in order to increase treatment efficacy. Interestingly, these ‘repositioned’ agents can target cancer cells, the tumor vasculature or, more broadly, the tumor microenvironment. Malignant tumors are no longer regarded as simple congregations of cancer cells but as genuine tissues with various components such as blood vessels, fibroblasts, inflammatory cells and an extracellular matrix. These different components and their multiple interactions play a crucial role in tumor development and response to treatment. Therefore, future anticancer treatments will have to take into account the tumor microenvironment and aim to target the different cellular and molecular participants encompassed in a tumor, as well as their specific interactions. In this article, we explain why MSAT represents a very attractive strategy for developing next-generation multitarget therapies.
Collapse
Affiliation(s)
- Nicolas André
- INSERM-UMR 911, Cytosquelette et Intégration des Signaux du Micro-Environnement Tumoral, CRO2, Université d’Aix-Marseille, Marseille, France; Hematology & Pediatric Oncology Department, Children Hospital of ‘La Timone’, AP-HM, Bd Jean Moulin 13885, Marseille Cedex 5, France
| | - Laetitia Padovani
- Service de Radiothérapie, Hôpital pour Enfants de ‘La Timone’, AP-HM, Marseille, France
| | - Eddy Pasquier
- Children’s Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Randwick, NSW, Australia
| |
Collapse
|
123
|
Liu CC, Lu N, Cui Y, Yang T, Zhao ZQ, Xin WJ, Liu XG. Prevention of paclitaxel-induced allodynia by minocycline: Effect on loss of peripheral nerve fibers and infiltration of macrophages in rats. Mol Pain 2010; 6:76. [PMID: 21050491 PMCID: PMC2991291 DOI: 10.1186/1744-8069-6-76] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/05/2010] [Indexed: 01/19/2023] Open
Abstract
Background Although paclitaxel is a frontline antineoplastic agent for treatment of solid tumors, the paclitaxel-evoked pain syndrome is a serious problem for patients. There is currently no valid drug to prevent or treat the paclitaxel-induced allodynia, partly due to lack of understanding regarding the cellular mechanism. Studies have shown that minocycline, an inhibitor of microglia/macrophage, prevented neuropathic pain and promoted neuronal survival in animal models of neurodegenerative disease. Recently, Cata et al also reported that minocycline inhibited allodynia induced by low-dose paclitaxel (2 mg/kg) in rats, but the mechanism is still unclear. Results Here, we investigate by immunohistochemistry the change of intraepidermal nerve fiber (IENF) in the hind paw glabrous skin, expression of macrophage and activating transcription factor 3 (ATF3) in DRG at different time points after moderate-dose paclitaxel treatment (cumulative dose 24 mg/kg; 3 × 8 mg/kg) in rats. Moreover, we observe the effect of minocycline on the IENF, macrophages and ATF3. The results showed that moderate-dose paclitaxel induced a persisted, gradual mechanical allodynia, which was accompanied by the loss of IENF in the hind paw glabrous skin and up-regulation of macrophages and ATF3 in DRG in rats. The expressions of ATF3 mainly focus on the NF200-positive cells. More importantly, we observed that pretreatment of minocycline at dose of 30 mg/kg or 50 mg/kg, but not 5 mg/kg, prevented paclitaxel-evoked allodynia. The evidence from immunohistochemistry showed that 30 mg/kg minocycline rescued the degeneration of IENF, attenuated infiltration of macrophages and up-regulation of ATF3 induced by paclitaxel treatment in rats. Conclusions Minocycline prevents paclitaxel-evoked allodynia, likely due to its inhibition on loss of IENF, infiltration of macrophages and up-regulation of ATF3 in rats. The finding might provide potential target for preventing paclitaxel-induced neuropathic pain.
Collapse
Affiliation(s)
- Cui-Cui Liu
- Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-Sen University, 74 Zhongshan Rd, 2, Guangzhou, 510080, PR China
| | | | | | | | | | | | | |
Collapse
|
124
|
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
|
125
|
Muralikrishnan G, Dinda AK, Shakeel F. Immunomodulatory Effects ofWithania Somniferaon Azoxymethane Induced Experimental Colon Cancer in Mice. Immunol Invest 2010; 39:688-98. [PMID: 20840055 DOI: 10.3109/08820139.2010.487083] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|