551
|
Papanikolaou J, Platogiannis N, Platogiannis D. Intrapericardial Cisplatin Instillation in Recurrent Postinfarction Cardiac Tamponade. J Cardiothorac Vasc Anesth 2017; 32:458-460. [PMID: 28939322 DOI: 10.1053/j.jvca.2017.04.041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Indexed: 11/11/2022]
Affiliation(s)
- John Papanikolaou
- Department of Cardiology, General Hospital of Trikala, Trikala, Thessaly, Greece; Department of Critical Care, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Thessaly, Greece.
| | | | | |
Collapse
|
552
|
Lian Q, Xu J, Yan S, Huang M, Ding H, Sun X, Bi A, Ding J, Sun B, Geng M. Chemotherapy-induced intestinal inflammatory responses are mediated by exosome secretion of double-strand DNA via AIM2 inflammasome activation. Cell Res 2017; 27:784-800. [PMID: 28409562 DOI: 10.1038/cr.2017.54] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/19/2017] [Accepted: 02/28/2017] [Indexed: 12/13/2022] Open
Abstract
Chemotherapies are known often to induce severe gastrointestinal tract toxicity but the underlying mechanism remains unclear. This study considers the widely applied cytotoxic agent irinotecan (CPT-11) as a representative agent and demonstrates that treatment induces massive release of double-strand DNA from the intestine that accounts for the dose-limiting intestinal toxicity of the compound. Specifically, "self-DNA" released through exosome secretion enters the cytosol of innate immune cells and activates the AIM2 (absent in melanoma 2) inflammasome. This leads to mature IL-1β and IL-18 secretion and induces intestinal mucositis and late-onset diarrhoea. Interestingly, abrogation of AIM2 signalling, either in AIM2-deficient mice or by a pharmacological inhibitor such as thalidomide, significantly reduces the incidence of drug-induced diarrhoea without affecting the anticancer efficacy of CPT-11. These findings provide mechanistic insights into how chemotherapy triggers innate immune responses causing intestinal toxicity, and reveal new chemotherapy regimens that maintain anti-tumour effects but circumvent the associated adverse inflammatory response.
Collapse
Affiliation(s)
- Qiaoshi Lian
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Xu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shanshan Yan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.,School of Life Sciences, University of Science and Technology of China, Hefei 230022, China
| | - Min Huang
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Honghua Ding
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaoyu Sun
- CAS Key Laboratory of Molecular Virology &Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Aiwei Bi
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Ding
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Bing Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.,CAS Key Laboratory of Molecular Virology &Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Meiyu Geng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
553
|
Wagner LM, Adams VR. Targeting the PD-1 pathway in pediatric solid tumors and brain tumors. Onco Targets Ther 2017; 10:2097-2106. [PMID: 28442918 PMCID: PMC5396947 DOI: 10.2147/ott.s124008] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
While remarkable advances have been made in the treatment of pediatric leukemia over the past decades, new therapies are needed for children with advanced solid tumors and high-grade brain tumors who fail standard chemotherapy regimens. Immunotherapy with immune checkpoint inhibitors acting through the programmed cell death-1 (PD-1) pathway has shown efficacy in some chemotherapy-resistant adult cancers, generating interest that these agents may also be helpful to treat certain refractory pediatric malignancies. In this manuscript we review current strategies for targeting the PD-1 pathway, highlighting putative biomarkers and the rationale for investigation of these drugs to treat common pediatric tumors such as sarcoma, neuroblastoma, and high-grade glioma. We summarize the completed and ongoing clinical trial data available, and suggest potential applications for further study.
Collapse
Affiliation(s)
| | - Val R Adams
- Department of Pharmacy Practice and Science, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
554
|
Topoisomerase II Inhibitors Induce DNA Damage-Dependent Interferon Responses Circumventing Ebola Virus Immune Evasion. mBio 2017; 8:mBio.00368-17. [PMID: 28377530 PMCID: PMC5380843 DOI: 10.1128/mbio.00368-17] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ebola virus (EBOV) protein VP35 inhibits production of interferon alpha/beta (IFN) by blocking RIG-I-like receptor signaling pathways, thereby promoting virus replication and pathogenesis. A high-throughput screening assay, developed to identify compounds that either inhibit or bypass VP35 IFN-antagonist function, identified five DNA intercalators as reproducible hits from a library of bioactive compounds. Four, including doxorubicin and daunorubicin, are anthracycline antibiotics that inhibit topoisomerase II and are used clinically as chemotherapeutic drugs. These compounds were demonstrated to induce IFN responses in an ATM kinase-dependent manner and to also trigger the DNA-sensing cGAS-STING pathway of IFN induction. These compounds also suppress EBOV replication in vitro and induce IFN in the presence of IFN-antagonist proteins from multiple negative-sense RNA viruses. These findings provide new insights into signaling pathways activated by important chemotherapy drugs and identify a novel therapeutic approach for IFN induction that may be exploited to inhibit RNA virus replication. Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses.
Collapse
|
555
|
Tevis KM, Cecchi RJ, Colson YL, Grinstaff MW. Mimicking the tumor microenvironment to regulate macrophage phenotype and assessing chemotherapeutic efficacy in embedded cancer cell/macrophage spheroid models. Acta Biomater 2017; 50:271-279. [PMID: 28011141 PMCID: PMC5316313 DOI: 10.1016/j.actbio.2016.12.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/30/2016] [Accepted: 12/19/2016] [Indexed: 01/02/2023]
Abstract
Tumor associated macrophages (TAMs) are critical stromal components intimately involved with the progression, invasion, and metastasis of cancer cells. To address the need for an in vitro system that mimics the clinical observations of TAM localizations and subsequent functional performance, a cancer cell/macrophage spheroid model is described. The central component of the model is a triple negative breast cancer spheroid embedded in a three-dimensional collagen gel. Macrophages are incorporated in two different ways. The first is a heterospheroid, a spheroid containing both tumor cells and macrophages. The heterospheroid mimics the population of TAMs infiltrated into the tumor mass, thus being exposed to hypoxia and metabolic gradients. In the second model, macrophages are diffusely seeded in the collagen surrounding the spheroid, thus modeling TAMs in the cancer stroma. The inclusion of macrophages as a heterospheroid changes the metabolic profile, indicative of synergistic growth. In contrast, macrophages diffusely seeded in the collagen bear the same profile regardless of the presence of a tumor cell spheroid. The macrophages in the heterospheroid secrete EGF, a cytokine critical to tumor/macrophage co-migration, and an EGF inhibitor decreases the metabolic activity of the heterospheroid, which is not observed in the other systems. The increased secretion of IL-10 indicates that the heterospheroid macrophages follow an M2/TAM differentiation pathway. Lastly, the heterospheroid exhibits resistance to paclitaxel. In summary, the collagen embedded heterospheroid model promotes TAM-like characteristics, and will be of utility in cancer biology and drug discovery. STATEMENT OF SIGNIFICANCE Two in vitro collagen-embedded multicellular spheroid models are described that mimic the clinical observations of macrophage localization within a tumor. Incorporation of macrophages within a breast cancer spheroid emphasizes cell-cell interactions with subsequent differentiation toward a tumor-promoting TAM phenotype. In contrast, macrophages seeded around the tumor spheroid display decreased interaction with cancer cells and no indication of a TAM phenotype. Finally, the presence of macrophages in the heterospheroid increases resistance to paclitaxel. This study demonstrates that cell-cell interactions and 3D collagen matrix direct macrophage activity, and, thus, highlights the important role the local environment itself plays in macrophage behavior.
Collapse
Affiliation(s)
- Kristie M Tevis
- Department of Biomedical Engineering, Cummington Street, Boston University, Boston, MA 02215, United States
| | - Ryan J Cecchi
- Department of Biomedical Engineering, Cummington Street, Boston University, Boston, MA 02215, United States
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02215, United States.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Cummington Street, Boston University, Boston, MA 02215, United States; Department of Chemistry, Metcalf Center for Science and Engineering, Boston University, Boston, MA 02215, United States; Department of Medicine, 715 Albany Street, Boston University, Boston, MA 02118, United States.
| |
Collapse
|
556
|
Chemotherapy-associated Posterior Reversible Encephalopathy Syndrome: A Case Report and Review of the Literature. Neurologist 2017; 21:112-117. [PMID: 27801773 DOI: 10.1097/nrl.0000000000000105] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION There are increasing reports of posterior reversible encephalopathy syndrome (PRES) associated with the use of chemotherapeutic agents. Recognition of PRES is crucial given its reversibility with appropriate supportive management. We report a patient presenting with PRES after treatment with Rituximab, Cyclophosphamide, Hydroxydaunorubicin/Adriamycin, Oncovin/Vincristine, Prednisone (R-CHOP) and intrathecal methotrexate. We also perform a systematic review of the literature on chemotherapy-associated PRES. CASE REPORT A 72-year-old man with recently diagnosed diffuse large B-cell lymphoma became unresponsive 4 days after initiation of R-CHOP and intrathecal methotrexate. Brain magnetic resonance imaging showed interval development of occipital and temporal fluid attenuation inversion recovery hyperintensities consistent with PRES. The patient's blood pressure was aggressively controlled and he received 5 days of high-dose methylprednisone. He subsequently regained consciousness and his mental status gradually improved. Repeat magnetic resonance imaging showed interval resolution of the bilateral fluid attenuation inversion recovery hyperintensities. REVIEW SUMMARY We performed a systematic review of the literature and included a total of 70 unique cases involving chemotherapy-associated PRES. Platinum-containing drugs, Cyclophosphamide, Hydroxydaunorubicin/Adriamycin, Oncovin/Vincristine, Prednisone/R-CHOP regimens, and gemcitabine were the agents most commonly used in patients who developed suspected chemo-associated PRES. Median onset of symptoms occurred 8 days after chemotherapy. Hypertension was the most commonly reported risk factor associated with the development of chemotherapy-associated PRES. In most cases, PRES improved with supportive management alone within 2 weeks. CONCLUSIONS Chemotherapy-associated PRES is an increasingly encountered syndrome. Both neurologists and non-neurologists should be familiar with the most commonly implicated agents, symptoms, risk factors, and clinical course of chemotherapy-associated PRES, given its favorable prognosis with appropriate management.
Collapse
|
557
|
Metronomic chemotherapy and immunotherapy in cancer treatment. Cancer Lett 2017; 400:282-292. [PMID: 28189534 DOI: 10.1016/j.canlet.2017.01.040] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 12/14/2022]
Abstract
Systemic chemotherapy given at maximum tolerated doses (MTD) has been the mainstay of cancer treatment for more than half a century. In some chemosensitive diseases such as hematologic malignancies and solid tumors, MTD has led to complete remission and even cure. The combination of maintenance therapy and standard MTD also can generate good disease control; however, resistance to chemotherapy and disease metastasis still remain major obstacles to successful cancer treatment in the majority of advanced tumors. Metronomic chemotherapy, defined as frequent administration of chemotherapeutic agents at a non-toxic dose without extended rest periods, was originally designed to overcome drug resistance by shifting the therapeutic target from tumor cells to tumor endothelial cells. Metronomic chemotherapy also exerts anti-tumor effects on the immune system (immunomodulation) and tumor cells. The goal of immunotherapy is to enhance host anti-tumor immunities. Adding immunomodulators such as metronomic chemotherapy to immunotherapy can improve the clinical outcomes in a synergistic manner. Here, we review the anti-tumor mechanisms of metronomic chemotherapy and the preliminary research addressing the combination of immunotherapy and metronomic chemotherapy for cancer treatment in animal models and in clinical setting.
Collapse
|
558
|
Safety and immunogenicity of neoadjuvant treatment using WT1-immunotherapeutic in combination with standard therapy in patients with WT1-positive Stage II/III breast cancer: a randomized Phase I study. Breast Cancer Res Treat 2017; 162:479-488. [PMID: 28176175 PMCID: PMC5332485 DOI: 10.1007/s10549-017-4130-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/20/2017] [Indexed: 12/23/2022]
Abstract
Purpose This Phase I, multicenter, randomized study (ClinicalTrials.gov NCT01220128) evaluated the safety and immunogenicity of recombinant Wilms’ tumor 1 (WT1) protein combined with the immunostimulant AS15 (WT1-immunotherapeutic) as neoadjuvant therapy administered concurrently with standard treatments in WT1-positive breast cancer patients. Methods Patients were treated in 4 cohorts according to neoadjuvant treatment (A: post-menopausal, hormone receptor [HR]-positive patients receiving aromatase inhibitors; B: patients receiving chemotherapy; C: HER2-overexpressing patients on trastuzumab–chemotherapy combination; D: HR-positive/HER2-negative patients on chemotherapy). Patients (cohorts A–C) were randomized (2:1) to receive 6 or 8 doses of WT1-immunotherapeutic or placebo together with standard neoadjuvant treatment in a double-blind manner; cohort D patients received WT1-immunotherapeutic in an open manner. Safety was assessed throughout the study. WT1-specific antibodies were assessed pre- and post-vaccination. Results Sixty-two patients were randomized; 60 received ≥ one dose of WT1-immunotherapeutic. Two severe toxicities were reported: diarrhea (cohort C; also reported as a grade 3 serious adverse event) and decreased left ventricular ejection fraction (cohort B; also reported as a grade 2 adverse event). Post-dose 4 of WT1-immunotherapeutic, 10/10 patients from cohort A, 0/8 patients from cohort B, 6/11 patients from cohort C, and 2/3 patients from cohort D were humoral responders. The sponsor elected to close the trial prematurely. Conclusions Concurrent administration of WT1-immunotherapeutic and standard neoadjuvant therapy was well tolerated and induced WT1-specific antibodies in patients receiving neoadjuvant aromatase inhibitors. In patients on neoadjuvant chemotherapy or trastuzumab–chemotherapy combination, the humoral response was impaired or blunted, likely due to either co-administration of corticosteroids and/or the chemotherapies themselves. Electronic supplementary material The online version of this article (doi:10.1007/s10549-017-4130-y) contains supplementary material, which is available to authorized users.
Collapse
|
559
|
Is there still a role for cytotoxic chemotherapy after targeted therapy and immunotherapy in metastatic melanoma? A case report and literature review. CHINESE JOURNAL OF CANCER 2017; 36:10. [PMID: 28086948 PMCID: PMC5237156 DOI: 10.1186/s40880-017-0179-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
Metastatic melanoma has long been considered to have a very poor prognosis and to be chemo-resistant. However, a subgroup of patients with metastatic melanoma presents remarkable responses to chemotherapeutic agents, even in the absence of a response to modern targeted therapies and immunotherapies; accordingly, determining predictive biomarkers of the response to chemotherapies for metastatic melanoma remains a priority to guide treatment in these patients. We report a case study of a patient with B-Raf proto-oncogene serine/threonine kinase-mutated metastatic melanoma harbouring many genetic mutations. The patient did not respond to prior targeted therapies or immunotherapies but experienced a dramatic objective radiological and clinical response to subsequent dacarbazine-based chemotherapy. In the era of targeted therapies and immunotherapies for metastatic melanoma, cytotoxic chemotherapies may still represent an interesting therapeutic weapon in a well-defined subgroup of patients presenting with specific genetic and molecular features.
Collapse
|
560
|
Bu X, Yao Y, Li X. Immune Checkpoint Blockade in Breast Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:383-402. [PMID: 29282694 DOI: 10.1007/978-981-10-6020-5_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is emerging as the most promising novel strategy for cancer treatment. Cancer immunotherapy is broadly categorized into three forms: immune checkpoint modulation, adoptive cell transfer, and cancer vaccine. Immune checkpoint blockade is demonstrated as the most clinically effective treatment with low immune-related adverse events (irAE). Blockade of PD-1/PD-L1 and CTLA-4 has achieved remarkable success in treating various types of tumors, which sparks great interests in this therapeutic strategy and expands the role of immune checkpoint blockade in treating tumors including breast cancer. Based on the notable results obtained from clinical trials, the United States' Food and Drug Administration (FDA) has approved multiple CTLA-4 monoclonal antibodies as well as the PD-1/PD-L1 monoclonal antibodies for treatment of different types of tumors. The theories of immunoediting, T-cell exhaustions, and co-stimulatory/co-inhibitory pathways are immunological foundations for immune checkpoint blockade therapy. Breast cancers such as triple negative breast cancer and HER-2 negative breast cancer respond to immune checkpoint blockade therapy due to their high immunogenicity. PD-1/PD-L1 blockade has just received FDA approval as a standard cancer therapy for solid tumors such as breast cancer. Development of immune checkpoint blockade focuses on two directions: one is to identify proper biomarkers of immune checkpoint blockade in breast cancer, and the other is to combine therapies with PD-1/PD-L1 blockade antibodies to achieve optimal clinical outcomes.
Collapse
Affiliation(s)
- Xia Bu
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China.
| | - Yihui Yao
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| | - Xiaoyu Li
- Department of Hematology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| |
Collapse
|
561
|
Kalinski P, Talmadge JE. Tumor Immuno-Environment in Cancer Progression and Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:1-18. [PMID: 29275461 DOI: 10.1007/978-3-319-67577-0_1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The approvals of Provenge (Sipuleucel-T), Ipilimumab (Yervoy/anti-CTLA-4) and blockers of the PD-1 - PD-L1/PD-L2 pathway, such as nivolumab (Opdivo), pembrolizumab (Keytruda), or atezolizumab (Tecentriq), have established immunotherapy as a key component of comprehensive cancer care. Further, murine mechanistic studies and studies in immunocompromised patients have documented the critical role of immunity in effectiveness of radio- and chemotherapy. However, in addition to the ability of the immune system to control cancer progression, it can also promote tumor growth, via regulatory T cells (Tregs), myeloid-derived dendritic cells (MDSCs) and tumor associated macrophages (TAM), which can enhance survival of cancer cells directly or via the regulation of the tumor stroma.An increasing body of evidence supports a central role for the tumor microenvironment (TME) and the interactions between tumor stroma, infiltrating immune cells and cancer cells during the induction and effector phase of anti-cancer immunity, and the overall effectiveness of immunotherapy and other forms of cancer treatment. In this chapter, we discuss the roles of key TME components during tumor progression, metastatic process and cancer therapy-induced tumor regression, as well as opportunities for their modulation to enhance the overall therapeutic benefit.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Medicine and Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY, USA.
| | - James E Talmadge
- University of Nebraska Medical Center, 986495 Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
562
|
Hude I, Sasse S, Engert A, Bröckelmann PJ. The emerging role of immune checkpoint inhibition in malignant lymphoma. Haematologica 2017; 102:30-42. [PMID: 27884973 PMCID: PMC5210230 DOI: 10.3324/haematol.2016.150656] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022] Open
Abstract
To evade elimination by the host immune system, tumor cells commonly exploit physiological immune checkpoint pathways, restraining efficient anti-tumor immune cell function. Growing understanding of the complex dialog between tumor cells and their microenvironment contributed to the development of immune checkpoint inhibitors. This innovative strategy has demonstrated paradigm-shifting clinical activity in various malignancies. Antibodies targeting programmed death 1 and cytotoxic T-lymphocyte-associated protein-4 are also being investigated in lymphoid malignancies with varying levels of activity and a favorable toxicity profile. To date, evaluated only in the setting of relapsed or refractory disease, anti-programmed death 1 antibodies such as nivolumab and pembrolizumab show encouraging response rates particularly in classical Hodgkin lymphoma but also in follicular lymphoma and diffuse-large B-cell lymphoma. As the first immune checkpoint inhibitor in lymphoma, nivolumab was approved for the treatment of relapsed or refractory classical Hodgkin lymphoma by the Food and Drug Administration in May 2016. In this review, we assess the role of the pathways involved and potential rationale of checkpoint inhibition in various lymphoid malignancies. In addition to data from current clinical trials, immune-related side effects, potential limitations and future perspectives including promising combinatory approaches with immune checkpoint inhibition are discussed.
Collapse
Affiliation(s)
- Ida Hude
- Department of Internal Medicine, Division of Hematology, University Hospital Center Zagreb, Croatia
| | - Stephanie Sasse
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Andreas Engert
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine and German Hodgkin Study Group (GHSG), University Hospital of Cologne, Germany
| |
Collapse
|
563
|
Black M, Barsoum IB, Truesdell P, Cotechini T, Macdonald-Goodfellow SK, Petroff M, Siemens DR, Koti M, Craig AWB, Graham CH. Activation of the PD-1/PD-L1 immune checkpoint confers tumor cell chemoresistance associated with increased metastasis. Oncotarget 2016; 7:10557-67. [PMID: 26859684 PMCID: PMC4891140 DOI: 10.18632/oncotarget.7235] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 01/25/2016] [Indexed: 12/13/2022] Open
Abstract
The ability of tumor cells to avoid immune destruction (immune escape) as well as their acquired resistance to anti-cancer drugs constitute important barriers to the successful management of cancer. Interaction between the Programmed Death Ligand 1 (PD-L1) on the surface of tumor cells with the Programmed Death-1 (PD-1) receptor on cytotoxic T lymphocytes leads to inactivation of these immune effectors and, consequently, immune escape. Here we show that the PD-1/PD-L1 axis also leads to tumor cell resistance to conventional chemotherapeutic agents. Using a panel of PD-L1-expressing human and mouse breast and prostate cancer cell lines, we found that incubation of breast and prostate cancer cells in the presence of purified recombinant PD-1 resulted in resistance to doxorubicin and docetaxel as determined using clonogenic survival assays. Co-culture with PD-1-expressing Jurkat T cells also promoted chemoresistance and this was prevented by antibody blockade of either PD-L1 or PD-1 or by silencing of the PD-L1 gene. Moreover, inhibition of the PD-1/PD-L1 axis using anti-PD-1 antibody enhanced doxorubicin chemotherapy to inhibit metastasis in a syngeneic mammary orthotopic mouse model of metastatic breast cancer. To further investigate the mechanism of tumor cell survival advantage upon PD-L1 ligation, we show that exposure to rPD-1 promoted ERK and mTOR growth and survival pathways leading to increased cell proliferation. Overall, the findings of this study indicate that combinations of chemotherapy and immune checkpoint blockade may limit chemoresistance and progression to metastatic disease.
Collapse
Affiliation(s)
- Madison Black
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ivraym B Barsoum
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Margaret Petroff
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - D Robert Siemens
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada
| | - Madhuri Koti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Andrew W B Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Department of Urology, Queen's University, Kingston, Ontario, Canada.,Cancer Biology and Genetics, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| |
Collapse
|
564
|
Welters MJ, van der Sluis TC, van Meir H, Loof NM, van Ham VJ, van Duikeren S, Santegoets SJ, Arens R, de Kam ML, Cohen AF, van Poelgeest MI, Kenter GG, Kroep JR, Burggraaf J, Melief CJ, van der Burg SH. Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci Transl Med 2016; 8:334ra52. [PMID: 27075626 DOI: 10.1126/scitranslmed.aad8307] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/05/2016] [Indexed: 12/27/2022]
Abstract
Therapeutic vaccination with human papillomavirus type 16 synthetic long peptides (HPV16-SLPs) results in T cell-mediated regression of HPV16-induced premalignant lesions but fails to install clinically effective immunity in patients with HPV16-positive cervical cancer. We explored whether HPV16-SLP vaccination can be combined with standard carboplatin and paclitaxel chemotherapy to improve immunity and which time point would be optimal for vaccination. This was studied in the HPV16 E6/E7-positive TC-1 mouse tumor model and in patients with advanced cervical cancer. In mice and patients, the presence of a progressing tumor was associated with abnormal frequencies of circulating myeloid cells. Treatment of TC-1-bearing mice with chemotherapy and therapeutic vaccination resulted in superior survival and was directly related to a chemotherapy-mediated altered composition of the myeloid cell population in the blood and tumor. Chemotherapy had no effect on tumor-specific T cell responses. In advanced cervical cancer patients, carboplatin-paclitaxel also normalized the abnormal numbers of circulating myeloid cells, and this was associated with increased T cell reactivity to recall antigens. The effect was most pronounced starting 2 weeks after the second cycle of chemotherapy, providing an optimal immunological window for vaccination. This was validated with a single dose of HPV16-SLP vaccine given in this time window. The resulting proliferative HPV16-specific T cell responses were unusually strong and were retained after all cycles of chemotherapy. In conclusion, carboplatin-paclitaxel therapy fosters vigorous vaccine-induced T cell responses when vaccination is given after chemotherapy and has reset the tumor-induced abnormal myeloid cell composition to normal values.
Collapse
Affiliation(s)
- Marij J Welters
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Tetje C van der Sluis
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Hélène van Meir
- Department of Gynecology, Leiden University Medical Center, 2300 RC Leiden, Netherlands. Centre for Human Drug Research, 2333 CL Leiden, Netherlands
| | - Nikki M Loof
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Vanessa J van Ham
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Suzanne van Duikeren
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Saskia J Santegoets
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | | | - Adam F Cohen
- Centre for Human Drug Research, 2333 CL Leiden, Netherlands
| | | | - Gemma G Kenter
- Department of Gynecology, Academic Medical Center, 1100 DD Amsterdam, Netherlands
| | - Judith R Kroep
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands
| | | | - Cornelis J Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300 RC Leiden, Netherlands. ISA Pharmaceuticals, 2333 CH Leiden, Netherlands
| | - Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, 2300 RC Leiden, Netherlands.
| |
Collapse
|
565
|
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: 6.8] [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
|
566
|
Ku AW, Muhitch JB, Powers CA, Diehl M, Kim M, Fisher DT, Sharda AP, Clements VK, O'Loughlin K, Minderman H, Messmer MN, Ma J, Skitzki JJ, Steeber DA, Walcheck B, Ostrand-Rosenberg S, Abrams SI, Evans SS. Tumor-induced MDSC act via remote control to inhibit L-selectin-dependent adaptive immunity in lymph nodes. eLife 2016; 5. [PMID: 27929373 PMCID: PMC5199197 DOI: 10.7554/elife.17375] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 12/07/2016] [Indexed: 12/23/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) contribute to an immunosuppressive network that drives cancer escape by disabling T cell adaptive immunity. The prevailing view is that MDSC-mediated immunosuppression is restricted to tissues where MDSC co-mingle with T cells. Here we show that splenic or, unexpectedly, blood-borne MDSC execute far-reaching immune suppression by reducing expression of the L-selectin lymph node (LN) homing receptor on naïve T and B cells. MDSC-induced L-selectin loss occurs through a contact-dependent, post-transcriptional mechanism that is independent of the major L-selectin sheddase, ADAM17, but results in significant elevation of circulating L-selectin in tumor-bearing mice. Even moderate deficits in L-selectin expression disrupt T cell trafficking to distant LN. Furthermore, T cells preconditioned by MDSC have diminished responses to subsequent antigen exposure, which in conjunction with reduced trafficking, severely restricts antigen-driven expansion in widely-dispersed LN. These results establish novel mechanisms for MDSC-mediated immunosuppression that have unanticipated implications for systemic cancer immunity. DOI:http://dx.doi.org/10.7554/eLife.17375.001
Collapse
Affiliation(s)
- Amy W Ku
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States
| | - Jason B Muhitch
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States.,Department of Urology, Roswell Park Cancer Institute, Buffalo, United States
| | - Colin A Powers
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, United States
| | - Michael Diehl
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States
| | - Minhyung Kim
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, United States
| | - Daniel T Fisher
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States.,Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, United States
| | - Anand P Sharda
- Department of Urology, Roswell Park Cancer Institute, Buffalo, United States
| | - Virginia K Clements
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, United States
| | - Kieran O'Loughlin
- Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, United States
| | - Hans Minderman
- Flow and Image Cytometry, Roswell Park Cancer Institute, Buffalo, United States
| | - Michelle N Messmer
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States
| | - Jing Ma
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, United States
| | - Joseph J Skitzki
- Department of Surgical Oncology, Roswell Park Cancer Institute, Buffalo, United States
| | - Douglas A Steeber
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, United States
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, United States
| | - Suzanne Ostrand-Rosenberg
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, United States
| | - Scott I Abrams
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States
| | - Sharon S Evans
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, United States
| |
Collapse
|
567
|
Marcq E, Siozopoulou V, De Waele J, van Audenaerde J, Zwaenepoel K, Santermans E, Hens N, Pauwels P, van Meerbeeck JP, Smits ELJ. Prognostic and predictive aspects of the tumor immune microenvironment and immune checkpoints in malignant pleural mesothelioma. Oncoimmunology 2016; 6:e1261241. [PMID: 28197385 DOI: 10.1080/2162402x.2016.1261241] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/27/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with a poor prognosis and an increasing incidence, for which novel therapeutic strategies are urgently required. Since the immune system has been described to play a presumed role in the protection against MPM, characterization of its tumor immune microenvironment (TME) and immune checkpoints can identify new immunotherapeutic targets and their predictive and/or prognostic value. To characterize the TME and the immune checkpoint expression profile, we performed immunohistochemistry (IHC) on formalin-fixed paraffin embedded (FFPE) tissue sections from 54 MPM patients (40 at time of diagnosis; 14 treated with chemotherapy). We stained for PD-1, PD-L1, TIM-3, LAG-3, CD4, CD8, CD45RO, granzyme B, FoxP3 and CD68. Furthermore, we analyzed the relationship between the immunological parameters and survival, as well as response to chemotherapy. We found that TIM-3, PD-1 and PD-L1 were expressed on both immune and tumor cells. Strikingly, PD-1 and PD-L1 expression on tumor cells was only seen in unpretreated samples. No LAG-3 expression was observed. CD45RO expression in the stroma was an independent negative predictive factor for response on chemotherapy, while CD4 and TIM-3 expression in lymphoid aggregates were independent prognostic factors for better outcome. Our data propose TIM-3 as a promising new target in mesothelioma. Chemotherapy influences the expression of immune checkpoints and therefore further research on the best combination treatment schedule is required.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | - Vasiliki Siozopoulou
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | | | - Karen Zwaenepoel
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Eva Santermans
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University , Diepenbeek, Belgium
| | - Niel Hens
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium; Centre for Health Economics Research and Modeling Infectious Diseases, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Thoracic Oncology/MOCA, Antwerp University Hospital, Antwerp, Belgium
| | - Evelien L J Smits
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
568
|
Chen Y, Xia R, Huang Y, Zhao W, Li J, Zhang X, Wang P, Venkataramanan R, Fan J, Xie W, Ma X, Lu B, Li S. An immunostimulatory dual-functional nanocarrier that improves cancer immunochemotherapy. Nat Commun 2016; 7:13443. [PMID: 27819653 PMCID: PMC5103075 DOI: 10.1038/ncomms13443] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 10/05/2016] [Indexed: 01/05/2023] Open
Abstract
Immunochemotherapy combines a chemotherapeutic agent with an immune-modulating agent and represents an attractive approach to improve cancer therapy. However, the success of immunochemotherapy is hampered by the lack of a strategy to effectively co-deliver the two therapeutics to the tumours. Here we report the development of a dual-functional, immunostimulatory nanomicellar carrier that is based on a prodrug conjugate of PEG with NLG919, an indoleamine 2,3-dioxygenase (IDO) inhibitor currently used for reversing tumour immune suppression. An Fmoc group, an effective drug-interactive motif, is also introduced into the carrier to improve the drug loading capacity and formulation stability. We show that PEG2k-Fmoc-NLG alone is effective in enhancing T-cell immune responses and exhibits significant antitumour activity in vivo. More importantly, systemic delivery of paclitaxel (PTX) using the PEG2k-Fmoc-NLG nanocarrier leads to a significantly improved antitumour response in both breast cancer and melanoma mouse models. The use of immunostimulatory agents to enhance the efficacy of chemotherapy is a promising strategy in cancer therapy. Here, the authors report on a micellar nanoparticle that can effectively co-deliver chemo- and immunotherapeutics, resulting in an improved in vivo antitumour response.
Collapse
Affiliation(s)
- Yichao Chen
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Rui Xia
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Yixian Huang
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Wenchen Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Jiang Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Xiaolan Zhang
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Pengcheng Wang
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Jie Fan
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Wen Xie
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Xiaochao Ma
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Binfeng Lu
- University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Song Li
- Center for Pharmacogenetics, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA.,University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| |
Collapse
|
569
|
Hsu FT, Chen TC, Chuang HY, Chang YF, Hwang JJ. Enhancement of adoptive T cell transfer with single low dose pretreatment of doxorubicin or paclitaxel in mice. Oncotarget 2016; 6:44134-50. [PMID: 26683520 PMCID: PMC4792547 DOI: 10.18632/oncotarget.6628] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 11/25/2015] [Indexed: 01/17/2023] Open
Abstract
Ex vivo expansion of CD8+ T-cells has been a hindrance for the success of adoptive T cell transfer in clinic. Currently, preconditioning with chemotherapy is used to modulate the patient immunity before ACT, however, the tumor microenvironment beneficial for transferring T cells may also be damaged. Here preconditioning with single low dose of doxorubicin or paclitaxel combined with fewer CD8+ T-cells was investigated to verify whether the same therapeutic efficacy of ACT could be achieved. An E.G7/OT1 animal model that involved adoptive transfer of OVA-specific CD8+ T-cells transduced with a granzyme B promoter-driven firefly luciferase and tomato fluorescent fusion reporter gene was used to evaluate this strategy. The result showed that CD8+ T-cells were activated and sustained longer in mice pretreated with one low-dose Dox or Tax. Enhanced therapeutic efficacy was found in Dox or Tax combined with 2x106 CD8+ T-cells and achieved the same level of tumor growth inhibition as that of 5x106 CD8+ T-cells group. Notably, reduced numbers of Tregs and myeloid derived suppressor cells were shown in combination groups. By contrast, the number of tumor-infiltrating cytotoxic T lymphocytes and IL-12 were increased. The NF-κB activity and immunosuppressive factors such as TGF-β, IDO, CCL2, VEGF, CCL22, COX-2 and IL-10 were suppressed. This study demonstrates that preconditioning with single low dose Dox or Tax and combined with two fifth of the original CD8+ T-cells could improve the tumor microenvironment via suppression of NF-κB and its related immunosuppressors, and activate more CD8+ T-cells which also stay longer.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan.,Translational Imaging Research Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Chun Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Fang Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Biophotonics and Molecular Imaging Research Center (BMIRC), National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
570
|
Grigg C, Blake Z, Gartrell R, Sacher A, Taback B, Saenger Y. Talimogene laherparepvec (T-Vec) for the treatment of melanoma and other cancers. Semin Oncol 2016; 43:638-646. [PMID: 28061981 DOI: 10.1053/j.seminoncol.2016.10.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/22/2016] [Indexed: 01/14/2023]
Abstract
Talimogene laherparepvec (T-Vec) is the first live virus to be approved by the US Food and Drug Administration for the treatment of cancer. This engineered version of herpes simplex virus type 1 (HSV-1) is the product of decades of preclinical work aimed at identifying and modifying aspects of the viral genome involved in virulence and immunogenicity. T-Vec preferentially infects and lyses tumor cells and, in some cases, induces a systemic immune response against the tumor. These properties have translated into significant and durable clinical responses, particularly in advanced melanoma. Many unanswered questions remain, including how to augment these clinical responses and which other tumor types may respond to oncolytic therapy. Here, we review the development of T-Vec, our current understanding of its impact on the tumor immune micro-environment, and its safety and efficacy in clinical trials for melanoma and other cancers.
Collapse
Affiliation(s)
- Claud Grigg
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Zoë Blake
- Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - Robyn Gartrell
- Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - Adrian Sacher
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Bret Taback
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY
| | - Yvonne Saenger
- Hematology/Oncology, New York-Presbyterian/Columbia University Medical Center, New York, NY.
| |
Collapse
|
571
|
Molecular Changes During Acute Myeloid Leukemia (AML) Evolution and Identification of Novel Treatment Strategies Through Molecular Stratification. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:383-436. [PMID: 27865463 DOI: 10.1016/bs.pmbts.2016.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Acute myeloid leukemia (AML) is a hematopoietic malignancy characterized by impaired differentiation and uncontrollable proliferation of myeloid progenitor cells. Due to high relapse rates, overall survival for this rapidly progressing disease is poor. The significant challenge in AML treatment is disease heterogeneity stemming from variability in maturation state of leukemic cells of origin, genetic aberrations among patients, and existence of multiple disease clones within a single patient. Disease heterogeneity and the lack of biomarkers for drug sensitivity lie at the root of treatment failure as well as selective efficacy of AML chemotherapies and the emergence of drug resistance. Furthermore, standard-of-care treatment is aggressive, presenting significant tolerability concerns to the commonly advanced-age AML patient. In this review, we examine the concept and potential of molecular stratification, particularly with biologically relevant drug responses, in identifying low-toxicity precision therapeutic combinations and clinically relevant biomarkers for AML patient care as a way to overcome these challenges in AML treatment.
Collapse
|
572
|
Hama Y. Exacerbation of gemcitabine-related pneumonia during radiotherapy for extrapulmonary lesion. Int Cancer Conf J 2016; 6:35-37. [PMID: 31149466 DOI: 10.1007/s13691-016-0267-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/29/2016] [Indexed: 11/25/2022] Open
Abstract
Late-onset gemcitabine pulmonary toxicity is rare and association between pulmonary toxicity and radiotherapy to the extrapulmonary sites is controversial. Here, we report a case of acute exacerbated fatal interstitial pneumonia during radiotherapy to the extrapulmonary site. A 73-year-old woman with pelvic lymph node metastases from urothelial carcinoma underwent palliative radiotherapy after failure of gemcitabine-containing and gemcitabine-non-containing chemotherapy. Gemcitabine-containing chemotherapy had finished 13 months prior to the radiotherapy due to grade 3 pulmonary toxicity. During the course of radiotherapy to the pelvic lesion, she was complicated with fatal acute interstitial pneumonia even though the lung tissue was not irradiated. To the best of our knowledge, this is the first reported case of fatal gemcitabine-related pulmonary toxicity during radiotherapy for extrapulmonary lesion. Although the association between late-onset pulmonary toxicity and radiotherapy is controversial, caution should be paid to a patient with a history of gemcitabine-related pulmonary toxicity who will undergo radiotherapy even though the lung volume is not irradiated.
Collapse
Affiliation(s)
- Yukihiro Hama
- Tokyo-Edogawa Cancer Center, Edogawa Hospital, 2-24-18 Higashikoiwa, Edogawa-ku, Tokyo, 133-0052 Japan
| |
Collapse
|
573
|
Evangelista FCG, Bandeira MO, Silva GD, Silva MG, Andrade SN, Marques DR, Silva LM, Castro WV, Santos FV, Viana GHR, Villar JAFP, Sabino AP, Varotti FP. Synthesis and in vitro evaluation of novel triazole/azide chalcones. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1705-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
574
|
Liu J, Blake SJ, Yong MCR, Harjunpää H, Ngiow SF, Takeda K, Young A, O'Donnell JS, Allen S, Smyth MJ, Teng MWL. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov 2016; 6:1382-1399. [PMID: 27663893 DOI: 10.1158/2159-8290.cd-16-0577] [Citation(s) in RCA: 579] [Impact Index Per Article: 72.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 12/17/2022]
Abstract
Immunotherapy has recently entered a renaissance phase with the approval of multiple agents for the treatment of cancer. Immunotherapy stands ready to join traditional modalities, including surgery, chemotherapy, radiation, and hormone therapy, as a pillar of cancer treatment. Although immunotherapy has begun to have success in advanced cancer treatment, its scheduling and efficacy with surgery to treat earlier stages of cancer and prevent distant metastases have not been systematically examined. Here, we have used two models of spontaneously metastatic breast cancers in mice to illustrate the significantly greater therapeutic power of neoadjuvant, compared with adjuvant, immunotherapies in the context of primary tumor resection. Elevated and sustained peripheral tumor-specific immune responses underpinned the outcome, and blood sampling of tumor-specific CD8+ T cells immediately prior to and post surgery may provide a predictor of outcome. These data now provide a strong rationale to extensively test and compare neoadjuvant immunotherapy in humans. SIGNIFICANCE We demonstrate the significantly greater therapeutic efficacy of neoadjuvant, compared with adjuvant, immunotherapies to eradicate distant metastases following primary tumor resection. Elevated and sustained peripheral tumor-specific immune responses underpinned the outcome, and blood sampling of tumor-specific CD8+ T cells immediately prior to and post surgery may provide a predictor of outcome. Cancer Discov; 6(12); 1382-99. ©2016 AACR.See related commentary by Melero et al., p. 1312This article is highlighted in the In This Issue feature, p. 1293.
Collapse
Affiliation(s)
- Jing Liu
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Stephen J Blake
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michelle C R Yong
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Heidi Harjunpää
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Shin Foong Ngiow
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, Japan
| | - Arabella Young
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Jake S O'Donnell
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Stacey Allen
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.,School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Michele W L Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. .,School of Medicine, University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
575
|
Schettini F, Giuliano M, De Placido S, Arpino G. Nab-paclitaxel for the treatment of triple-negative breast cancer: Rationale, clinical data and future perspectives. Cancer Treat Rev 2016; 50:129-141. [PMID: 27665540 DOI: 10.1016/j.ctrv.2016.09.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/14/2016] [Accepted: 09/01/2016] [Indexed: 01/05/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for ∼10-20% of breast cancers and is associated with relatively poor prognosis, earlier disease recurrence and higher number of visceral metastases. Despite an increasing understanding of the molecular heterogeneity of TNBC, clinical trials of targeted agents have thus far been disappointing; chemotherapy, in particular with anthracycline and taxanes, remains the backbone medical management for both early and metastatic TNBC. Nab-paclitaxel is a solvent-free, albumin-bound, nanoparticle formulation of paclitaxel and represents a novel formulation of an established, effective chemotherapeutic agent. Nab-paclitaxel has been specifically designed to overcome the limitations of conventional taxane formulations, including the barriers to effective drug delivery of highly lipophilic agents. It has shown significant efficacy and better tolerability than conventional taxanes in metastatic breast cancer and is approved for use in this setting. Increasing evidence suggests that nab-paclitaxel is effective in patients with more aggressive tumours, as seen in TNBC. Indeed, results of Phase II/III studies indicate that nab-paclitaxel may be effective as neoadjuvant treatment of TNBC. This article reviews the rationale and evidence supporting a role for nab-paclitaxel in the treatment of TNBC, including ongoing studies such as ADAPT-TN and tnAcity. In addition, the article reviews ongoing research into targeted therapies and immuno-oncology for the treatment of TNBC, and explores the potential role, current evidence and ongoing studies of nab-paclitaxel as the chemotherapy partner in combination with immunotherapy, where the unique properties of this taxane, including the lack of requirement for steroid pre-medication, may present an advantage.
Collapse
Affiliation(s)
- Francesco Schettini
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| | - Mario Giuliano
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy; Lester and Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, 77030 Houston, TX, USA.
| | - Sabino De Placido
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| | - Grazia Arpino
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
576
|
Plattel WJ, Alsada ZND, van Imhoff GW, Diepstra A, van den Berg A, Visser L. Biomarkers for evaluation of treatment response in classical Hodgkin lymphoma: comparison of sGalectin-1, sCD163 and sCD30 with TARC. Br J Haematol 2016; 175:868-875. [PMID: 27610595 DOI: 10.1111/bjh.14317] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/13/2016] [Indexed: 01/03/2023]
Abstract
Soluble Galectin-1 (sGal-1, also termed LGALS1), soluble CD163 (sCD163) and soluble CD30 (sCD30) have been reported to be elevated in plasma or serum of patients with classical Hodgkin lymphoma (cHL). We aimed to determine the clinical utility of these biomarkers for evaluation of treatment response compared to thymus and activation regulated chemokine (TARC, also termed CCL17). Plasma or serum samples were prospectively collected among 103 newly diagnosed cHL patients before and after treatment. Levels of sGal-1, sCD163, sCD30 and TARC were correlated with disease characteristics and clinical treatment response. Elevated plasma levels of sGal-1, sCD163, sCD30 and TARC were found in 67%, 21%, 91% and 93% of cHL patients respectively. Mean plasma levels of sGal-1 and sCD30 decreased after treatment but sCD163 did not decrease after treatment. There was no correlation with change of these markers and clinical treatment response in individual patients. TARC levels strongly correlated with disease characteristics and metabolic volume. TARC remained high in 6 out of 7 non-responsive patients and dramatically decreased in 95 out of 96 responsive patients. In summary, elevated pre-treatment levels of sGal-1, sCD163, sCD30 and TARC can be found in patients with cHL. However, only plasma TARC accurately reflects disease activity and correlates with clinical treatment response.
Collapse
Affiliation(s)
- Wouter J Plattel
- Department of Haematology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Zainab N D Alsada
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Gustaaf W van Imhoff
- Department of Haematology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Lydia Visser
- Department of Pathology and Medical Biology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
577
|
Tiffen JC, Gallagher SJ, Tseng HY, Filipp FV, Fazekas de St. Groth B, Hersey P. EZH2 as a mediator of treatment resistance in melanoma. Pigment Cell Melanoma Res 2016; 29:500-7. [PMID: 27063195 PMCID: PMC5021620 DOI: 10.1111/pcmr.12481] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 03/24/2016] [Indexed: 12/27/2022]
Abstract
Direct treatments of cancer such as chemotherapy, radiotherapy and targeted therapy have been shown to depend on recruitment of the immune system for their effectiveness. Recent studies have shown that development of resistance to direct therapies such as BRAF inhibitors in melanoma is associated with suppression of immune responses. We point to emerging data that implicate activation of the polycomb repressive complex 2 (PRC2) and its catalytic component-enhancer of zeste homolog 2 (EZH2)-in progression of melanoma and suppression of immune responses. EZH2 appears to have an important role in differentiation of CD4 T cells and particularly in the function of T regulatory cells, which suppress immune responses to melanoma. We review mechanisms of EZH2 activation at the genomic level and from activation of the MAP kinase, E2F or NF-kB2 pathways. These studies are consistent with activation of EZH2 as a common mechanism for induction of immune suppression in patients failing direct therapies and suggest EZH2 inhibitors may have a role in combination with immunotherapy and targeted therapies to prevent development of immunosuppression.
Collapse
Affiliation(s)
- Jessamy C Tiffen
- Melanoma Immunology and Oncology Group, The Centenary Institute, University of Sydney, Camperdown, NSW, Australia
| | - Stuart J Gallagher
- Melanoma Immunology and Oncology Group, The Centenary Institute, University of Sydney, Camperdown, NSW, Australia
| | - Hsin-Yi Tseng
- Melanoma Immunology and Oncology Group, The Centenary Institute, University of Sydney, Camperdown, NSW, Australia
| | - Fabian V Filipp
- Systems Biology and Cancer Metabolism, Program for Quantitative Systems Biology, University of California Merced, Merced, CA, USA
| | | | - Peter Hersey
- Melanoma Immunology and Oncology Group, The Centenary Institute, University of Sydney, Camperdown, NSW, Australia.
| |
Collapse
|
578
|
Uhle F, Chousterman BG, Grützmann R, Brenner T, Weber GF. Pathogenic, immunologic, and clinical aspects of sepsis - update 2016. Expert Rev Anti Infect Ther 2016; 14:917-27. [PMID: 27530423 DOI: 10.1080/14787210.2016.1224971] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Sepsis is a major cause of death worldwide but its orchestrating components remain incompletely understood. On the one hand, development of sepsis results from an infectious focus that cannot be controlled by the immune system, but on the other, responding immune cells that can eliminate the infection inflict damage to the host by contributing to complications such as endothelial leakage, septic shock, and multiorgan failure. AREAS COVERED In this review we give a comprehensive overview of how sepsis occurs, which exogenous and endogenous factors might affect the immune-pathophysiological course of sepsis and finally how this knowledge translates into up-to-date definitions and therapeutic approaches. Expert commentary: Although new immunological mechanisms altering the course of sepsis have been identified recently, future research needs to address the limitations of experimental approaches, redirect the research focus into translational approaches, and finally evaluate personalized treatment strategies.
Collapse
Affiliation(s)
- Florian Uhle
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Benjamin G Chousterman
- b Department of Anesthesia, Intensive Care and SAMU , Hôpital Lariboisière, AP-HP, and Université Paris Diderot , Paris , France
| | - Robert Grützmann
- c Department of Surgery , University Hospital Erlangen-Nürnberg , Erlangen , Germany
| | - Thorsten Brenner
- a Department of Anesthesiology , Heidelberg University Hospital , Heidelberg , Germany
| | - Georg F Weber
- c Department of Surgery , University Hospital Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
579
|
Yue T, Zheng X, Dou Y, Zheng X, Sun R, Tian Z, Wei H. Interleukin 12 shows a better curative effect on lung cancer than paclitaxel and cisplatin doublet chemotherapy. BMC Cancer 2016; 16:665. [PMID: 27549240 PMCID: PMC4994391 DOI: 10.1186/s12885-016-2701-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 08/10/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Interleukin 12 (IL-12) is a cytokine that has been reported to exhibit potent tumoricidal effects in animal tumor models. A combined approach using Paclitaxel and platinum-based doublet chemotherapy is the most commonly used backbone regimen for treating lung cancer. Despite numerous studies regarding the anti-tumor effects of IL-12 and the widespread use of conventional chemotherapy, few direct comparisons of IL-12 and conventional chemotherapy in the treatment of lung cancer have been performed. METHODS We compared IL-12 to paclitaxel and cisplatin doublet chemotherapy in terms of efficacy against lung cancer in mouse models. The antitumor effect was measured by survival assays, histological analyses and imaging analyses. The cytokine levels were assessed using enzyme linked immunosorbent assay (ELISA) and flow cytometry (FACS). The spleen sizes were measured. CD31, CD105 and Vascular endothelial growth factor receptor 3 (VEGFR3) were analyzed using immunofluorescence. Matrix metalloprotein-9 (MMP-9) and cadherin 1 (CDH1) transcript levels were measured by quantitative PCR. Tumor cells apoptosis were examined by Tunel assay. RESULTS The results showed that IL-12 treatment inhibited lung tumor growth, resulting in the long-term survival of lung cancer-bearing mice. Further examination revealed that IL-12 rapidly activated NK cells to secrete IFN-γ, resulting in the inhibition of tumor angiogenesis. In contrast, paclitaxel and cisplatin doublet chemotherapy did not show the expected efficacy in orthotopic lung cancer models; the IFN-γ levels were not increased after this treatment, and the number of peripheral lymphocytes was reduced. CONCLUSION Together, these animal model data indicate that IL-12 shows a better curative effect than PTX + CDDP doublet chemotherapy.
Collapse
Affiliation(s)
- Ting Yue
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaodong Zheng
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Yaling Dou
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaohu Zheng
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Sun
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China.,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhigang Tian
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. .,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.
| | - Haiming Wei
- Institute of Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China. .,Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
580
|
Grigg C, Rizvi NA. PD-L1 biomarker testing for non-small cell lung cancer: truth or fiction? J Immunother Cancer 2016; 4:48. [PMID: 27532023 PMCID: PMC4986262 DOI: 10.1186/s40425-016-0153-x] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023] Open
Abstract
Research in cancer immunology is currently accelerating following a series of cancer immunotherapy breakthroughs during the last 5 years. Various monoclonal antibodies which block the interaction between checkpoint molecules PD-1 on immune cells and PD-L1 on cancer cells have been used to successfully treat non-small cell lung cancer (NSCLC), including some durable responses lasting years. Two drugs, nivolumab and pembrolizumab, are now FDA approved for use in certain patients who have failed or progressed on platinum-based or targeted therapies while agents targeting PD-L1, atezolizumab and durvalumab, are approaching the final stages of clinical testing. Despite impressive treatment outcomes in a subset of patients who receive these immune therapies, many patients with NSCLC fail to respond to anti-PD-1/PD-L1 and the identification of a biomarker to select these patients remains highly sought after. In this review, we discuss the recent clinical trial results of pembrolizumab, nivolumab, and atezolizumab for NSCLC, and the significance of companion diagnostic testing for tumor PD-L1 expression.
Collapse
Affiliation(s)
- Claud Grigg
- NewYork-Presbyterian/Columbia University Medical Center, Hematology/Oncology, 177 Fort Washington Avenue, 6GN-435, New York, NY 10032 USA
| | - Naiyer A Rizvi
- NewYork-Presbyterian/Columbia University Medical Center, Hematology/Oncology, 177 Fort Washington Avenue, 6GN-435, New York, NY 10032 USA
| |
Collapse
|
581
|
Lu Y, Wang Y, Miao L, Haynes M, Xiang G, Huang L. Exploiting in situ antigen generation and immune modulation to enhance chemotherapy response in advanced melanoma: A combination nanomedicine approach. Cancer Lett 2016; 379:32-8. [DOI: 10.1016/j.canlet.2016.05.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 02/25/2016] [Accepted: 05/21/2016] [Indexed: 01/30/2023]
|
582
|
Di Biase S, Lee C, Brandhorst S, Manes B, Buono R, Cheng CW, Cacciottolo M, Martin-Montalvo A, de Cabo R, Wei M, Morgan TE, Longo VD. Fasting-Mimicking Diet Reduces HO-1 to Promote T Cell-Mediated Tumor Cytotoxicity. Cancer Cell 2016; 30:136-146. [PMID: 27411588 PMCID: PMC5388544 DOI: 10.1016/j.ccell.2016.06.005] [Citation(s) in RCA: 277] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 01/30/2016] [Accepted: 06/10/2016] [Indexed: 12/22/2022]
Abstract
Immune-based interventions are promising strategies to achieve long-term cancer-free survival. Fasting was previously shown to differentially sensitize tumors to chemotherapy while protecting normal cells, including hematopoietic stem and immune cells, from its toxic side effects. Here, we show that the combination of chemotherapy and a fasting-mimicking diet (FMD) increases the levels of bone marrow common lymphoid progenitor cells and cytotoxic CD8(+) tumor-infiltrating lymphocytes (TILs), leading to a major delay in breast cancer and melanoma progression. In breast tumors, this effect is partially mediated by the downregulation of the stress-responsive enzyme heme oxygenase-1 (HO-1). These data indicate that FMD cycles combined with chemotherapy can enhance T cell-dependent targeted killing of cancer cells both by stimulating the hematopoietic system and by enhancing CD8(+)-dependent tumor cytotoxicity.
Collapse
Affiliation(s)
- Stefano Di Biase
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Changhan Lee
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Sebastian Brandhorst
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Brianna Manes
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Roberta Buono
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Chia-Wei Cheng
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Mafalda Cacciottolo
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Alejandro Martin-Montalvo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Min Wei
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Todd E. Morgan
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D. Longo
- Longevity Institute, Leonard Davis School of Gerontology and Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
- IFOM, FIRC Institute of Molecular Oncology, Milano, Italy
- Correspondence to:
| |
Collapse
|
583
|
Antibody-Drug Conjugates for Cancer Therapy. Biomedicines 2016; 4:biomedicines4030014. [PMID: 28536381 PMCID: PMC5344263 DOI: 10.3390/biomedicines4030014] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/24/2016] [Accepted: 06/27/2016] [Indexed: 11/18/2022] Open
Abstract
Antibody–drug conjugates (ADCs) take advantage of the specificity of a monoclonal antibody to deliver a linked cytotoxic agent directly into a tumour cell. The development of these compounds provides exciting opportunities for improvements in patient care. Here, we review the key issues impacting on the clinical success of ADCs in cancer therapy. Like many other developing therapeutic classes, there remain challenges in the design and optimisation of these compounds. As the clinical applications for ADCs continue to expand, key strategies to improve patient outcomes include better patient selection for treatment and the identification of mechanisms of therapy resistance.
Collapse
|
584
|
Economopoulou P, Agelaki S, Perisanidis C, Giotakis EI, Psyrri A. The promise of immunotherapy in head and neck squamous cell carcinoma. Ann Oncol 2016; 27:1675-85. [PMID: 27380958 DOI: 10.1093/annonc/mdw226] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 05/29/2016] [Indexed: 01/05/2023] Open
Abstract
Squamous cell cancers of the head and neck (HNSCC) comprise a diverse group of malignancies that includes tobacco-related tumors in addition to an increasing number of human papillomavirus-associated cancers. Independently of cause, there is a growing body of evidence supporting that the immune system plays a pivotal role in HNSCC development, as tumor cells evade immunosurveillance by exploiting inhibitory checkpoint pathways that suppress anti-tumor T-cell responses. HNSCC cells have the ability to manipulate the immune system through a variety of different mechanisms, forcing it to promote tumor growth and spread. Over the last decade, discoveries in immunologic research resulted in increased understanding of complex interactions between HNSCC and the host immune system as well as T-cell regulatory mechanisms, promoting the development of a variety of novel immunotherapies. Following the availability of novel immunotherapeutic strategies, the challenge for clinicians is to understand how and in which clinical setting to use these agents in order to provide greater clinical benefit for patients. Combination of immunotherapies with standard treatment approaches also represents an evolving field of research. Herein, we provide a comprehensive review of immune escape mechanisms in HNSCC, as well as current immunotherapy approaches under investigation.
Collapse
Affiliation(s)
- P Economopoulou
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, School of Medicine, Athens
| | - S Agelaki
- Department of Medical Oncology, University Hospital of Heraklion, Heraklion Laboratory of Tumor Biology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - C Perisanidis
- Department of Maxillofacial and Oral Surgery, Medical University of Vienna, Vienna, Austria
| | - E I Giotakis
- Department of Otorhinolaryngology, Facial Plastic and Reconstructive Surgery, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany
| | - A Psyrri
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, National Kapodistrian University of Athens, School of Medicine, Athens
| |
Collapse
|
585
|
Hoeller C, Michielin O, Ascierto PA, Szabo Z, Blank CU. Systematic review of the use of granulocyte-macrophage colony-stimulating factor in patients with advanced melanoma. Cancer Immunol Immunother 2016; 65:1015-34. [PMID: 27372293 PMCID: PMC4995227 DOI: 10.1007/s00262-016-1860-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 06/15/2016] [Indexed: 12/24/2022]
Abstract
Several immunomodulatory checkpoint inhibitors have been approved for the treatment of patients with advanced melanoma, including ipilimumab, nivolumab and pembrolizumab. Talimogene laherparepvec is the first oncolytic virus to gain regulatory approval in the USA; it is also approved in Europe. Talimogene laherparepvec expresses granulocyte–macrophage colony-stimulating factor (GM-CSF), and with other GM-CSF-expressing oncolytic viruses in development, understanding the clinical relevance of this cytokine in treating advanced melanoma is important. Results of trials of GM-CSF in melanoma have been mixed, and while GM-CSF has the potential to promote anti-tumor responses, some preclinical data suggest that GM-CSF may sometimes promote tumor growth. GM-CSF has not been approved as a melanoma treatment. We undertook a systematic literature review of studies of GM-CSF in patients with advanced melanoma (stage IIIB–IV). Of the 503 articles identified, 26 studies met the eligibility criteria. Most studies investigated the use of GM-CSF in combination with another treatment, such as peptide vaccines or chemotherapy, or as an adjuvant to surgery. Some clinical benefit was reported in patients who received GM-CSF as an adjuvant to surgery, or in combination with other treatments. In general, outcomes for patients receiving peptide vaccines were not improved with the addition of GM-CSF. GM-CSF may be a valuable therapeutic adjuvant; however, further studies are needed, particularly head-to-head comparisons, to confirm the optimal dosing regimen and clinical effectiveness in patients with advanced melanoma.
Collapse
Affiliation(s)
- Christoph Hoeller
- Department of Dermatology, Medical University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Olivier Michielin
- Department of Oncology, Lausanne University Hospital, Champ de l'Air, Rue du Bugnon 21, 1011, Lausanne, Switzerland.,Ludwig Centre and Swiss Institute of Bioinformatics, Génopode Building, 1015, Lausanne, Switzerland
| | - Paolo A Ascierto
- Istituto Nazionale Tumori, Fondazione 'G. Pascale', Via Mariano Semmola, 52, 80131, Naples, Italy
| | - Zsolt Szabo
- Clinical Development, Amgen Europe GmbH, Dammstrasse 23, 6300, Zug, Switzerland
| | - Christian U Blank
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands
| |
Collapse
|
586
|
Cheng WL, Kao YH, Chen SA, Chen YJ. Pathophysiology of cancer therapy-provoked atrial fibrillation. Int J Cardiol 2016; 219:186-94. [PMID: 27327505 DOI: 10.1016/j.ijcard.2016.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/11/2016] [Indexed: 02/07/2023]
Abstract
Atrial fibrillation (AF) occurs with increased frequency in cancer patients, especially in patients who undergo surgery or chemotherapy. AF disturbs the prognosis of cancer patients and challenges therapeutic outcomes of cancer treatment. Elucidating the mechanisms of cancer-induced AF would help identify specific strategies for preventing AF occurrence. In addition to concurrent risk factors of cancer and AF, cancer surgery, side effects of anticancer agents, and cancer-associated immune responses play critical roles in the genesis of AF. In this review, we provide succinct potential mechanisms of AF genesis in cancer patients.
Collapse
Affiliation(s)
- Wan-Li Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ann Chen
- School of Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology and Cardiovascular Research Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
587
|
Immune-system-dependent anti-tumor activity of a plant-derived polyphenol rich fraction in a melanoma mouse model. Cell Death Dis 2016; 7:e2243. [PMID: 27253407 PMCID: PMC5143373 DOI: 10.1038/cddis.2016.134] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/29/2016] [Accepted: 03/30/2016] [Indexed: 12/22/2022]
Abstract
Recent findings suggest that part of the anti-tumor effects of several chemotherapeutic agents require an intact immune system. This is in part due to the induction of immunogenic cell death. We have identified a gallotannin-rich fraction, obtained from Caesalpinia spinosa (P2Et) as an anti-tumor agent in both breast carcinoma and melanoma. Here, we report that P2Et treatment results in activation of caspase 3 and 9, mobilization of cytochrome c and externalization of annexin V in tumor cells, thus suggesting the induction of apoptosis. This was preceded by the onset of autophagy and the expression of immunogenic cell death markers. We further demonstrate that P2Et-treated tumor cells are highly immunogenic in vaccinated mice and induce immune system activation, clearly shown by the generation of interferon gamma (IFN-γ) producing tyrosine-related protein 2 antigen-specific CD8+ T cells. Moreover, the tumor protective effects of P2Et treatment were abolished in immunodeficient mice, and partially lost after CD4 and CD8 depletion, indicating that P2Et's anti-tumor activity is highly dependent on immune system and at least in part of T cells. Altogether, these results support the hypothesis that the gallotannin-rich fraction P2Et's anti-tumor effects are mediated to a great extent by the endogenous immune response following to the exposure to immunogenic dying tumor cells.
Collapse
|
588
|
Noble F, Mellows T, McCormick Matthews LH, Bateman AC, Harris S, Underwood TJ, Byrne JP, Bailey IS, Sharland DM, Kelly JJ, Primrose JN, Sahota SS, Bateman AR, Thomas GJ, Ottensmeier CH. Tumour infiltrating lymphocytes correlate with improved survival in patients with oesophageal adenocarcinoma. Cancer Immunol Immunother 2016; 65:651-62. [PMID: 27020682 PMCID: PMC4880639 DOI: 10.1007/s00262-016-1826-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 03/08/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Oesophageal adenocarcinoma (OAC) is increasingly common in the west, and survival remains poor at 10-15 % at 5 years. Immune responses are increasingly implicated as a determining factor of tumour progression. The ability of lymphocytes to recognise tumour antigens provides a mechanism for a host immune attack against cancer providing a potential treatment strategy. MATERIALS AND METHODS Tumour infiltrating lymphocytes (TILs: CD3+, CD4+, CD8+ and FOXp3+) were assessed by immunohistochemistry using tissue microarrays in a contemporary and homogeneous cohort of OAC patients (n = 128) undergoing curative treatment. RESULTS Multivariate analysis identified three independent prognostic factors for improved cancer-specific survival (CSS): increased CD8+ TILs (p = 0.003), completeness of resection (p < 0.0001) and lower pathological N stage (p < 0.0001). Independent prognostic factors for favourable disease-free survival included surgery-only treatment (p = 0.015), completeness of resection (p = 0.001), increased CD8+ TILs (p < 0.0001) and reduced pathological N stage (p < 0.0001). Higher levels of TILs in the pathological specimen were associated with significant pathological response to neoadjuvant chemotherapy (NAC). On multivariate analysis increased levels of CD4+ (p = 0.017) and CD8+ TILs (p = 0.005) were associated with significant local tumour regression and lymph node downstaging, respectively. DISCUSSION Our results establish an association of TILs and survival in OAC, as seen in other solid tumours, and identify particular TIL subsets that are present at higher levels in patients who responded to NAC compared to non-responders. These findings highlight potential therapeutic strategies in EAC based on utilising the host immunological response and highlight the immune responses biomarker potential.
Collapse
Affiliation(s)
- Fergus Noble
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK.
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK.
| | - Toby Mellows
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
| | - Leo H McCormick Matthews
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
| | - Adrian C Bateman
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Scott Harris
- Public Health Sciences and Medical Statistics, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Timothy J Underwood
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James P Byrne
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Ian S Bailey
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Donna M Sharland
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Jamie J Kelly
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - John N Primrose
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Surinder S Sahota
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
| | - Andrew R Bateman
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
- Cancer Care, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Gareth J Thomas
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christian H Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building (MP824), Southampton General Hospital, Tremona Road, Southampton, UK
- Cancer Care, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
589
|
New developments for antibody-drug conjugate-based therapeutic approaches. Curr Opin Immunol 2016; 40:14-23. [DOI: 10.1016/j.coi.2016.02.008] [Citation(s) in RCA: 195] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 01/05/2023]
|
590
|
Vasundhara M, Baranwal M, Kumar A. Fusarium tricinctum, An Endophytic Fungus Exhibits Cell Growth Inhibition and Antioxidant Activity. Indian J Microbiol 2016; 56:433-438. [PMID: 27784939 DOI: 10.1007/s12088-016-0600-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022] Open
Abstract
An endophytic fungus (strain T6) isolated from Taxus baccata was studied for its effect on the growth of human breast cancer cell line (MCF-7), human cervical cancer cell line (HeLa) and peripheral blood mononuclear cells (PBMCs) as well as for its antioxidant activity. Based on morphological characters and internal transcribed spacer (ITS) sequence analysis, this fungus (strain T6) was identified as Fusarium tricinctum. This fungus has shown inhibition in the growth of the MCF-7 and HeLa cancer cell lines. IC50 values of the fungal extract were 225 ± 26 and 220 ± 18 μg ml-1 for MCF-7 and HeLa cell lines, respectively. Further, F. tricinctum showed inhibition in the proliferation of concanavalin A stimulated PBMCs indicating its immunosuppressive potential (IC50 value 110 ± 44 μg ml-1). Tumour necrosis factor (TNF)-α production in concanavalin A stimulated PBMCs and MCF-7 were found to be inhibited which indicates that the antiproliferative effect may be associated with TNF-α. Free radical scavenging results revealed that this fungus also exhibited antioxidant activity (IC50 value 482 ± 9 μg ml-1). Present study results suggested that F. tricinctum has the potential to be used for therapeutic purposes because of its antiproliferative and antioxidant potential.
Collapse
Affiliation(s)
- M Vasundhara
- Department of Biotechnology, Thapar University, Patiala, Punjab 147004 India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar University, Patiala, Punjab 147004 India
| | - Anil Kumar
- Department of Biotechnology, Thapar University, Patiala, Punjab 147004 India
| |
Collapse
|
591
|
de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ 2016; 23:733-47. [PMID: 26943322 PMCID: PMC4832109 DOI: 10.1038/cdd.2015.174] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.
Collapse
Affiliation(s)
- D de Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - J Lemke
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - A Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - H Walczak
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - L Martinez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón, Zaragoza, Spain
| |
Collapse
|
592
|
Lerner SP, Bajorin DF, Dinney CP, Efstathiou JA, Groshen S, Hahn NM, Hansel D, Kwiatkowski D, O’Donnell M, Rosenberg J, Svatek R, Abrams JS, Al-Ahmadie H, Apolo AB, Bellmunt J, Callahan M, Cha EK, Drake C, Jarow J, Kamat A, Kim W, Knowles M, Mann B, Marchionni L, McConkey D, McShane L, Ramirez N, Sharabi A, Sharpe AH, Solit D, Tangen CM, Amiri AT, Van Allen E, West PJ, Witjes JA, Quale DZ. Summary and Recommendations from the National Cancer Institute's Clinical Trials Planning Meeting on Novel Therapeutics for Non-Muscle Invasive Bladder Cancer. Bladder Cancer 2016; 2:165-202. [PMID: 27376138 PMCID: PMC4927845 DOI: 10.3233/blc-160053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The NCI Bladder Cancer Task Force convened a Clinical Trials Planning Meeting (CTPM) Workshop focused on Novel Therapeutics for Non-Muscle Invasive Bladder Cancer (NMIBC). Meeting attendees included a broad and multi-disciplinary group of clinical and research stakeholders and included leaders from NCI, FDA, National Clinical Trials Network (NCTN), advocacy and the pharmaceutical and biotech industry. The meeting goals and objectives were to: 1) create a collaborative environment in which the greater bladder research community can pursue future optimally designed novel clinical trials focused on the theme of molecular targeted and immune-based therapies in NMIBC; 2) frame the clinical and translational questions that are of highest priority; and 3) develop two clinical trial designs focusing on immunotherapy and molecular targeted therapy. Despite successful development and implementation of large Phase II and Phase III trials in bladder and upper urinary tract cancers, there are no active and accruing trials in the NMIBC space within the NCTN. Disappointingly, there has been only one new FDA approved drug (Valrubicin) in any bladder cancer disease state since 1998. Although genomic-based data for bladder cancer are increasingly available, translating these discoveries into practice changing treatment is still to come. Recently, major efforts in defining the genomic characteristics of NMIBC have been achieved. Aligned with these data is the growing number of targeted therapy agents approved and/or in development in other organ site cancers and the multiple similarities of bladder cancer with molecular subtypes in these other cancers. Additionally, although bladder cancer is one of the more immunogenic tumors, some tumors have the ability to attenuate or eliminate host immune responses. Two trial concepts emerged from the meeting including a window of opportunity trial (Phase 0) testing an FGFR3 inhibitor and a second multi-arm multi-stage trial testing combinations of BCG or radiotherapy and immunomodulatory agents in patients who recur after induction BCG (BCG failure).
Collapse
Affiliation(s)
| | - Dean F. Bajorin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Colin P. Dinney
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Susan Groshen
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Noah M. Hahn
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Donna Hansel
- University of California, La Jolla, San Diego, CA, USA
| | - David Kwiatkowski
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jonathan Rosenberg
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Robert Svatek
- UT Health Science Center San Antonio, San Antonio, TX, USA
| | - Jeffrey S. Abrams
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joaquim Bellmunt
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Margaret Callahan
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Eugene K. Cha
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles Drake
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Jonathan Jarow
- Office of Hematology and Oncology Products, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ashish Kamat
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - William Kim
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Margaret Knowles
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Bhupinder Mann
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luigi Marchionni
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - David McConkey
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lisa McShane
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Nilsa Ramirez
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Andrew Sharabi
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Arlene H. Sharpe
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - David Solit
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College of Cornell University, New York, NY, USA
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Eliezer Van Allen
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | | | - J. A. Witjes
- Department of Urology, Radboud UMC, Nijmegen, The Netherlands
| | | |
Collapse
|
593
|
Shaked Y. Balancing efficacy of and host immune responses to cancer therapy: the yin and yang effects. Nat Rev Clin Oncol 2016; 13:611-26. [PMID: 27118493 DOI: 10.1038/nrclinonc.2016.57] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Local and systemic treatments for cancer include surgery, radiation, chemotherapy, hormonal therapy, molecularly targeted therapies, antiangiogenic therapy, and immunotherapy. Many of these therapies can be curative in patients with early stage disease, but much less frequently is this the case when they are used to treat advanced-stage metastatic disease. In the latter setting, innate and/or acquired resistance are among the reasons for reduced responsiveness or nonresponsiveness to therapy, or for tumour relapse after an initial response. Most studies of resistance or reduced responsiveness focus on 'driver' genetic (or epigenetic) changes in the tumour-cell population. Several studies have highlighted the contribution of therapy-induced physiological changes in host tissues and cells that can reduce or even nullify the desired antitumour effects of therapy. These unwanted host effects can promote tumour-cell proliferation (repopulation) and even malignant aggressiveness. These effects occur as a result of systemic release of numerous cytokines, and mobilization of various host accessory cells, which can invade the treated tumour microenvironment. In short, the desired tumour-targeting effects of therapy (the 'yin') can be offset by a reactive host response (the 'yang'); proactively preventing or actively suppressing the latter represents a possible new approach to improving the efficacy of both local and systemic cancer therapies.
Collapse
Affiliation(s)
- Yuval Shaked
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, 1 Efron St. Bat Galim, Haifa 31096, Israel
| |
Collapse
|
594
|
Hylander BL, Repasky EA. Thermoneutrality, Mice, and Cancer: A Heated Opinion. Trends Cancer 2016; 2:166-175. [PMID: 28741570 DOI: 10.1016/j.trecan.2016.03.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/31/2022]
Abstract
The 'mild' cold stress caused by standard sub-thermoneutral housing temperatures used for laboratory mice in research institutes is sufficient to significantly bias conclusions drawn from murine models of several human diseases. We review the data leading to this conclusion, discuss the implications for research and suggest ways to reduce problems in reproducibility and experimental transparency caused by this housing variable. We have found that these cool temperatures suppress endogenous immune responses, skewing tumor growth data and the severity of graft versus host disease, and also increase the therapeutic resistance of tumors. Owing to the potential for ambient temperature to affect energy homeostasis as well as adrenergic stress, both of which could contribute to biased outcomes in murine cancer models, housing temperature should be reported in all publications and considered as a potential source of variability in results between laboratories. Researchers and regulatory agencies should work together to determine whether changes in housing parameters would enhance the use of mouse models in cancer research, as well as for other diseases. Finally, for many years agencies such as the National Cancer Institute (NCI) have encouraged the development of newer and more sophisticated mouse models for cancer research, but we believe that, without an appreciation of how basic murine physiology is affected by ambient temperature, even data from these models is likely to be compromised.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Roswell Park Cancer Institute, Department of Immunology, Elm and Carlton Streets, Buffalo, NY 14263-0001, USA
| | - Elizabeth A Repasky
- Roswell Park Cancer Institute, Department of Immunology, Elm and Carlton Streets, Buffalo, NY 14263-0001, USA.
| |
Collapse
|
595
|
Carotta S. Targeting NK Cells for Anticancer Immunotherapy: Clinical and Preclinical Approaches. Front Immunol 2016; 7:152. [PMID: 27148271 PMCID: PMC4838611 DOI: 10.3389/fimmu.2016.00152] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 04/07/2016] [Indexed: 11/23/2022] Open
Abstract
The recent success of checkpoint blockade has highlighted the potential of immunotherapy approaches for cancer treatment. Although the majority of approved immunotherapy drugs target T cell subsets, it is appreciated that other components of the immune system have important roles in tumor immune surveillance as well and thus represent promising additional targets for immunotherapy. Natural killer (NK) cells are the body’s first line of defense against infected or transformed cells, as they kill target cells in an antigen-independent manner. Although several studies have clearly demonstrated the active role of NK cells in cancer immune surveillance, only few clinically approved therapies currently exist that harness their potential. Our increased understanding of NK cell biology over the past few years has renewed the interest in NK cell-based anticancer therapies, which has lead to a steady increase of NK cell-based clinical and preclinical trials. Here, the role of NK cells in cancer immune surveillance is summarized, and several novel approaches to enhance NK cell cytotoxicity against cancer are discussed.
Collapse
Affiliation(s)
- Sebastian Carotta
- Immune Modulation Department, Boehringer Ingelheim RCV, Vienna, Austria; The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| |
Collapse
|
596
|
Cheng Q, Liu Y. Multifunctional platinum-based nanoparticles for biomedical applications. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [DOI: 10.1002/wnan.1410] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 03/07/2016] [Accepted: 03/17/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Qinqin Cheng
- CAS Key Laboratory of Soft Matter Chemistry, CAS High Magnetic Field Laboratory, Department of Chemistry; University of Science and Technology of China; Hefei China
| | - Yangzhong Liu
- CAS Key Laboratory of Soft Matter Chemistry, CAS High Magnetic Field Laboratory, Department of Chemistry; University of Science and Technology of China; Hefei China
| |
Collapse
|
597
|
Koide H, Okamoto A, Tsuchida H, Ando H, Ariizumi S, Kiyokawa C, Hashimoto M, Asai T, Dewa T, Oku N. One-step encapsulation of siRNA between lipid-layers of multi-layer polycation liposomes by lipoplex freeze-thawing. J Control Release 2016; 228:1-8. [DOI: 10.1016/j.jconrel.2016.01.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/05/2016] [Accepted: 01/18/2016] [Indexed: 01/09/2023]
|
598
|
Epigenetics and innate immunity: the ‘unTolld’ story. Immunol Cell Biol 2016; 94:631-9. [DOI: 10.1038/icb.2016.24] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/18/2016] [Accepted: 02/19/2016] [Indexed: 12/19/2022]
|
599
|
Linares J, Fernández AB, Feito MJ, Matesanz MC, Sánchez-Salcedo S, Arcos D, Vallet-Regí M, Rojo JM, Portolés MT. Effects of nanocrystalline hydroxyapatites on macrophage polarization. J Mater Chem B 2016; 4:1951-1959. [PMID: 32263072 DOI: 10.1039/c6tb00014b] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Silicon substituted and nanocrystalline hydroxyapatites have attracted the attention of many researchers due to their up-regulation in osteoblast cell metabolism and enhanced bioreactivity, respectively. On the other hand, the biomaterial success or failure depends ultimately on the immune response triggered after its implantation. Macrophages are the main components of the innate immune system with an important role in healing and tissue remodelling due to their remarkable functional plasticity, existing in a whole spectrum of functional populations with varying phenotypic features. The effects of nanocrystalline hydroxyapatite (nano-HA) and nanocrystalline silicon substituted hydroxyapatite (nano-SiHA) on the macrophage populations defined as pro-inflammatory (M1) and reparative (M2) phenotypes have been evaluated in the present study using RAW 264.7 cells and mouse peritoneal macrophages as in vitro models. M1 and M2 macrophage phenotypes were characterized by flow cytometry and confocal microscopy by the expression of CD80 and CD163, known as M1 and M2 markers, respectively. The polarization of primary macrophages towards the M1 or M2 phenotype was induced with the pro-inflammatory stimulus LPS or the anti-inflammatory stimulus IL-10, respectively, evaluating the biomaterial effects under these conditions. Our results show that both nano-HA and nano-SiHA favour the macrophage polarization towards an M2 reparative phenotype, decreasing M1 population and ensuring an appropriate response in the implantation site of these biomaterials designed for bone repair and bone tissue engineering.
Collapse
Affiliation(s)
- J Linares
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry, Universidad Complutense de Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
600
|
Tagliamonte M, Petrizzo A, Napolitano M, Luciano A, Rea D, Barbieri A, Arra C, Maiolino P, Tornesello M, Ciliberto G, Buonaguro FM, Buonaguro L. A novel multi-drug metronomic chemotherapy significantly delays tumor growth in mice. J Transl Med 2016; 14:58. [PMID: 26911136 PMCID: PMC4766679 DOI: 10.1186/s12967-016-0812-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/09/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The tumor immunosuppressive microenvironment represents a major obstacle to an effective tumor-specific cellular immune response. METHODS In the present study, the counterbalance effect of a novel metronomic chemotherapy protocol on such an immunosuppressive microenvironment was evaluated in a mouse model upon sub-cutaneous ectopic implantation of B16 melanoma cells. The chemotherapy consisted of a novel multi-drug cocktail including taxanes and alkylating agents, administered in a daily metronomic fashion. The newly designed strategy was shown to be safe, well tolerated and significantly efficacious. RESULTS Treated animals showed a remarkable delay in tumor growth and prolonged survival as compared to control group. Such an effect was directly correlated with CD4(+) T cell reduction and CD8(+) T cell increase. Furthermore, a significant reduction in the percentage of both CD25(+)FoxP3(+) and CD25(+)CD127(low) regulatory T cell population was found both in the spleens and in the tumor lesions. Finally, the metronomic chemotherapy induced an intrinsic CD8(+) T cell response specific to B16 naturally expressed Trp2 TAA. CONCLUSION The novel multi-drug daily metronomic chemotherapy evaluated in the present study was very effective in counterbalancing the immunosuppressive tumor microenvironment. Consequently, the intrinsic anti-tumor T cell immunity could exert its function, targeting specific TAA and significantly containing tumor growth. Overall, the results show that this represents a promising adjuvant approach to significantly enhance efficacy of intrinsic or vaccine-elicited tumor-specific cellular immunity.
Collapse
Affiliation(s)
- Maria Tagliamonte
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Annacarmen Petrizzo
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Maria Napolitano
- Laboratory of Clinical Immunology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Antonio Luciano
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Domenica Rea
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Claudio Arra
- Animal Facility, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Marialina Tornesello
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Gennaro Ciliberto
- Scientific Direction, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Franco M Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| | - Luigi Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione Pascale" - IRCCS, Naples, Italy.
| |
Collapse
|