1
|
Yau JNN, Adriani G. Three-dimensional heterotypic colorectal cancer spheroid models for evaluation of drug response. Front Oncol 2023; 13:1148930. [PMID: 37469395 PMCID: PMC10352797 DOI: 10.3389/fonc.2023.1148930] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/15/2023] [Indexed: 07/21/2023] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide. Improved preclinical tumor models are needed to make treatment screening clinically relevant and address disease mortality. Advancements in 3D cell culture have enabled a greater recapitulation of the architecture and heterogeneity of the tumor microenvironment (TME). This has enhanced their pathophysiological relevance and enabled more accurate predictions of tumor progression and drug response in patients. An increasing number of 3D CRC spheroid models include cell populations such as cancer-associated fibroblasts (CAFs), endothelial cells (ECs), immune cells, and gut bacteria to better mimic the in vivo regulation of signaling pathways. Furthermore, cell heterogeneity within the 3D spheroid models enables the identification of new therapeutic targets to develop alternative treatments and test TME-target therapies. In this mini review, we present the advances in mimicking tumor heterogeneity in 3D CRC spheroid models by incorporating CAFs, ECs, immune cells, and gut bacteria. We introduce how, in these models, the diverse cells influence chemoresistance and tumor progression of the CRC spheroids. We also highlight important parameters evaluated during drug screening in the CRC heterocellular spheroids.
Collapse
Affiliation(s)
- Jia Ning Nicolette Yau
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Rimini M, Puzzoni M, Pedica F, Silvestris N, Fornaro L, Aprile G, Loi E, Brunetti O, Vivaldi C, Simionato F, Zavattari P, Scartozzi M, Burgio V, Ratti F, Aldrighetti L, Cascinu S, Casadei-Gardini A. Cholangiocarcinoma: new perspectives for new horizons. Expert Rev Gastroenterol Hepatol 2021; 15:1367-1383. [PMID: 34669536 DOI: 10.1080/17474124.2021.1991313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Biliary tract cancer represents a heterogeneous group of malignancies characterized by dismal prognosis and scarce therapeutic options. AREA COVERED In the last years, a growing interest in BTC pathology has emerged, thus highlighting a significant heterogeneity of the pathways underlying the carcinogenesis process, from both a molecular and genomic point of view. A better understanding of these differences is mandatory to deepen the behavior of this complex disease, as well as to identify new targetable target mutations, with the aim to improve the survival outcomes. The authors decided to provide a comprehensive overview of the recent highlights on BTCs, with a special focus on the genetic, epigenetic and molecular alterations, which may have an interesting clinical application in the next future. EXPERT OPINION In the last years, the efforts resulted from international collaborations have led to the identification of new promising targets for precision medicine approaches in the BTC setting. Further investigations and prospective trials are needed, but the hope is that these new knowledge in cooperation with the new technologies and procedures, including bio-molecular and genomic analysis as well radiomic studies, will enrich the therapeutic armamentarium thus improving the survival outcomes in a such lethal and complex disease.
Collapse
Affiliation(s)
- Margherita Rimini
- Department of Oncology and Hematology, Division of Oncology, University of Modena and Reggio Emilia, Modena, Italy
| | - Marco Puzzoni
- Medical Oncology, University and University Hospital of Cagliari, Italy
| | - Federica Pedica
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Silvestris
- Department of oncology, Instituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy.,Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Lorenzo Fornaro
- Department of medical oncology, U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Eleonora Loi
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, Cagliari, Italy
| | - Oronzo Brunetti
- Department of oncology, Instituto Di Ricovero E Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Caterina Vivaldi
- Department of medical oncology, U.O. Oncologia Medica 2 Universitaria, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Francesca Simionato
- Department of Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Patrizia Zavattari
- Department of Biomedical Sciences, Unit of Biology and Genetics, University of Cagliari, Cagliari, Italy
| | - Mario Scartozzi
- Medical Oncology, University and University Hospital of Cagliari, Italy
| | - Valentina Burgio
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Francesca Ratti
- Hepatobiliary Surgery Division, IRCCS San Raffaele and Vita-Salute University, Italy
| | - Luca Aldrighetti
- Hepatobiliary Surgery Division, IRCCS San Raffaele and Vita-Salute University, Italy
| | - Stefano Cascinu
- Department of Oncology, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | | |
Collapse
|
3
|
Lampis A, Ratti M, Ghidini M, Mirchev MB, Okuducu AF, Valeri N, Hahne JC. Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). Int J Mol Med 2021; 47:97. [PMID: 33846775 PMCID: PMC8041478 DOI: 10.3892/ijmm.2021.4930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Oesophageal cancer is one of the most aggressive malignancies with limited treatment options, thus resulting in a high morbidity and mortality. With 5‑year survival rates of only 5‑10%, oesophageal cancer holds a dismal prognosis for patients. In order to improve overall survival, the early diagnosis and tools for patient stratification for personalized treatment are urgent needs. A minority of oesophageal cancers belong to the spectrum of Lynch syndrome‑associated cancers and are characterized by microsatellite instability (MSI). Microsatellite instability is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumours, such as colorectal and gastric cancer. In the latter, high levels of MSI are associated with a better prognosis and with an increased benefit to immune‑based therapies. Therefore, similar therapeutic approaches could offer an opportunity of treatment for oesophageal cancer patients with MSI. Apart from immune checkpoint inhibitors, other immunotherapies such as adoptive T‑cell transfer, peptide vaccine and oncolytic viruses are under investigation in oesophageal cancer patients. In the present review, the rationale and current knowledge about immunotherapies in oesophageal cancer are summarised.
Collapse
Affiliation(s)
- Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| | - Margherita Ratti
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Medical Department, Division of Oncology, Hospital Trust of Cremona, I-26100 Cremona, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Hospital Policlinic 'Fondazione IRCCS Ca' Granda Ospedale Maggiore', I-20122 Milan, Italy
| | - Milko B. Mirchev
- Clinic of Gastroenterology, Medical University, 9002 Varna, Bulgaria
| | | | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, Sutton SM25NG, UK
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| |
Collapse
|
4
|
Wang W, Wu M, Liu M, Yan Z, Wang G, Mao D, Wang M. Hyperprogression to camrelizumab in a patient with esophageal squamous cell carcinoma harboring EGFR kinase domain duplication. J Immunother Cancer 2020; 8:jitc-2020-000793. [PMID: 32581041 PMCID: PMC7312344 DOI: 10.1136/jitc-2020-000793] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background Previous studies have reported that the amplification of some genes, such as Murine Double Minute 2 or 4 and Epidermal Growth Factor Receptor (EGFR), may be related to hyperprogressive disease (HPD). Exploring somatic gene alterations might be an effective method to predict HPD. Herein we characterize the somatic alterations in a patient with esophageal squamous cell carcinoma (ESCC) who developed HPD to investigate the potential origins of HPD. Case presentation A man in his mid-40s was diagnosed with ESCC. After the failure of first-line treatment with cisplatin and docetaxel, the patient participated in a phase III randomized, open, multicenter clinical trial (CTR20170307) and subsequently received camrelizumab. After 4 weeks of immunotherapy, the tumor size increased by 79% compared with baseline imaging; the progressive pace was 2.5-fold higher than preimmunotherapy, and a new liver metastasis appeared. A rare EGFR exon 2–28 duplication was discovered in both preimmunotherapy and postimmunotherapy tumor tissues. Conclusion This is the first report on a patient with ESCC harboring rare EGFR kinase domain duplication in exons 2–28 and developing HPD in the process of camrelizumab treatment. This case suggested that EGFR kinase domain duplication might be associated with HPD. Administration of immune checkpoint inhibitor monotherapy in this subgroup of patients harboring EGFR kinase domain duplication should be performed with caution. These results need to be further confirmed in a larger cohort of patients.
Collapse
Affiliation(s)
- Wei Wang
- Department of Oncology, Changhai Hospital of Shanghai, Shanghai, China
| | - Meihong Wu
- Department of Oncology, Changhai Hospital of Shanghai, Shanghai, China
| | - Minglu Liu
- Department of Radiology, Changhai Hospital of Shanghai, Shanghai, China
| | - Zhengqing Yan
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Guoqiang Wang
- The Medical Department, 3D Medicines Inc, Shanghai, China
| | - Dongliang Mao
- Department of Oncology, North Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mei Wang
- Department of Oncology, North Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
5
|
Musacchio L, Boccia SM, Caruso G, Santangelo G, Fischetti M, Tomao F, Perniola G, Palaia I, Muzii L, Pignata S, Benedetti Panici P, Di Donato V. Immune Checkpoint Inhibitors: A Promising Choice for Endometrial Cancer Patients? J Clin Med 2020; 9:jcm9061721. [PMID: 32503218 PMCID: PMC7356971 DOI: 10.3390/jcm9061721] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/25/2020] [Accepted: 05/29/2020] [Indexed: 01/05/2023] Open
Abstract
Although around 80% of endometrial cancers are diagnosed at early stages and present with a 5-year survival rate exceeding 95%, patients with advanced and recurrent disease show a poor prognosis and low response rates to standard chemotherapy. In the era of targeted therapy, the great advances in the understanding of programmed death-ligand 1 (PD-L1) upregulation in cancer cells, which is responsible for tumor immune escape, have contributed to the increasing interest in immune checkpoint inhibitors as a promising strategy for the treatment of several refractory solid malignancies, including endometrial cancer. Several clinical trials have investigated the efficacy and safety of immune checkpoint inhibitors in endometrial cancer, which already led to the approval of the anti-programmed cell death protein 1 (anti-PD-1) antibody pembrolizumab as a satisfactory alternative for selected patients with unresectable or metastatic disease. As the future of cancer treatment will probably rely on combination therapy strategies, currently, innovative ongoing trials are exploring the potential role of immune checkpoint inhibitors associated with chemotherapy, radiotherapy, and other targeted therapies. Moreover, further research is warranted to discover new specific biomarkers that can accurately predict the response to immunotherapy.
Collapse
Affiliation(s)
- Lucia Musacchio
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Serena Maria Boccia
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Giuseppe Caruso
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
- Correspondence: ; Tel.: +39-0649972535
| | - Giusi Santangelo
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Margherita Fischetti
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Federica Tomao
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Innocenza Palaia
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Ludovico Muzii
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, 80131 Naples, Italy;
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| | - Violante Di Donato
- Department of Maternal and Child Health and Urological Sciences, University of Rome “Sapienza”, Policlinico “Umberto I”, 00161 Rome, Italy; (L.M.); (S.M.B.); (G.S.); (M.F.); (F.T.); (G.P.); (I.P.); (L.M.); (P.B.P.); (V.D.D.)
| |
Collapse
|
6
|
Jones WD, Michener CM, Biscotti C, Braicu I, Sehouli J, Ganapathi MK, Ganapathi RN. RNA Immune Signatures from Pan-Cancer Analysis Are Prognostic for High-Grade Serous Ovarian Cancer and Other Female Cancers. Cancers (Basel) 2020; 12:cancers12030620. [PMID: 32156016 PMCID: PMC7139955 DOI: 10.3390/cancers12030620] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/03/2020] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Immune cell infiltrates within the tumor microenvironment can influence treatment response and outcome in several cancers. In this study, we developed RNA-based immune signatures from pan-cancer analysis that could serve as potential markers across tumor types and tested them for association with outcome in high-grade serous ovarian cancer (HGSOC) and other female cancers. Pan-cancer RNA-Seq cluster analysis of immune-related gene expression profiles in The Cancer Genome Atlas (TCGA) from 29 different solid tumors (4446 specimens) identified distinct but concordant gene signatures. Among these immune signatures, Cytotoxic Lymphocyte Immune Signature (CLIS), T-cell trafficking (TCT), and the TCT to M2 tumor-associated macrophage (M2TAM) ratio (TCT:M2TAM) were significantly (p < 0.05) associated with overall survival (OS), using multivariable Cox proportional hazards regression models, in a discovery cohort and two independent validation cohorts of HGSOC patients. Notably, the TCT:M2TAM ratio was highly significant (p ≤ 0.000001) in two HGSOC cohorts. Immune signatures were also significant (p < 0.05) in the presence of tumor cytoreduction, BRCA1/2 mutation, and COL2A1 expression. Importantly, the CLIS and TCT signatures were also validated for prognostic significance (p < 0.05) in TCGA cohorts for endometrial and high tumor mutational burden (Hi-TMB) breast cancer. These immune signatures also have the potential for being predictive in other cancers and for patients following different treatment strategies.
Collapse
Affiliation(s)
- Wendell D. Jones
- Bioinformatics Group, Q Solutions - EA Genomics, 5927 S Miami Blvd, Morrisville, NC 27560, USA;
| | - Chad M. Michener
- Division of Gynecologic Oncology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA;
| | - Charles Biscotti
- Department of Anatomic Pathology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA;
| | - Iona Braicu
- Department of Gynecology, Charité Medical University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (I.B.); (J.S.)
| | - Jalid Sehouli
- Department of Gynecology, Charité Medical University of Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (I.B.); (J.S.)
| | - Mahrukh K. Ganapathi
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas Medical Center, 1021 Morehead Medical Drive, Charlotte, NC 28204, USA;
| | - Ram N. Ganapathi
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas Medical Center, 1021 Morehead Medical Drive, Charlotte, NC 28204, USA;
- Correspondence: or
| |
Collapse
|
7
|
Courau T, Bonnereau J, Chicoteau J, Bottois H, Remark R, Assante Miranda L, Toubert A, Blery M, Aparicio T, Allez M, Le Bourhis L. Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J Immunother Cancer 2019; 7:74. [PMID: 30871626 PMCID: PMC6417026 DOI: 10.1186/s40425-019-0553-9] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Background Immunotherapies still fail to benefit colorectal cancer (CRC) patients. Relevant functional assays aimed at studying these failures and the efficacy of cancer immunotherapy in human are scarce. 3D tumor cultures, called tumor organoids or spheroids, represent interesting models to study cancer treatments and could help to challenge these issues. Methods We analyzed heterotypic cocultures of human colon tumor-derived spheroids with immune cells to assess the infiltration, activation and function of T and NK cells toward human colorectal tumors in vitro. Results We showed that allogeneic T and NK cells rapidly infiltrated cell line-derived spheroids, inducing immune-mediated tumor cell apoptosis and spheroid destruction. NKG2D, a key activator of cytotoxic responses, was engaged on infiltrating cells. We thus assessed the therapeutic potential of an antibody targeting the specific ligands of NKG2D, MICA and MICB, in this system. Anti-MICA/B enhanced immune-dependent destruction of tumor spheroid by driving an increased NK cells infiltration and activation. Interestingly, tumor cells reacted to immune infiltration by upregulating HLA-E, ligand of the inhibitory receptor NKG2A expressed by CD8 and NK cells. NKG2A was increased after anti-MICA/B treatment and, accordingly, combination of anti-MICA/B and anti-NKG2A was synergistic. These observations were ultimately confirmed in a clinical relevant model of coculture between CRC patients-derived spheroids and autologous tumor-infiltrating lymphocytes. Conclusions Altogether, we show that tumor spheroids represent a relevant tool to study tumor-lymphocyte interactions on human tissues and revealed the antitumor potential of immunomodulatory antibodies targeting MICA/B and NKG2A. Electronic supplementary material The online version of this article (10.1186/s40425-019-0553-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tristan Courau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France
| | - Julie Bonnereau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | - Justine Chicoteau
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Hugo Bottois
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | | | | | - Antoine Toubert
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France
| | | | - Thomas Aparicio
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Matthieu Allez
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.,Paris-Diderot University, Sorbonne Paris Cité, Paris, France.,Gastroenterology and Digestive Oncology Department, Saint Louis Hospital, AP-HP, Paris, France
| | - Lionel Le Bourhis
- INSERM U1160, Institut de Recherche Saint-Louis, Saint Louis Hospital, Paris, France.
| |
Collapse
|
8
|
MHC Class 1 and PDL-1 Status of Primary Tumor and Lymph Node Metastatic Tumor Tissue in Gastric Cancers. Gastroenterol Res Pract 2019; 2019:4785098. [PMID: 30881447 PMCID: PMC6381579 DOI: 10.1155/2019/4785098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/08/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022] Open
Abstract
The prognosis of metastatic gastric cancer is poor. Despite the use of VEGF-, EGFR-, and HER2-targeting agents, prognosis is still poor in advanced gastric cancer. Although cancer immunotherapy responds well in some patients, clinical use is limited due to unanswered patients. For this reason, it is necessary to know the characteristics of primary and metastatic cancer cells for patient selection for immunotherapy and additional criteria are required. MHC-1 downregulation is most frequently observed in the tumor escape mechanism of cancer cells from the immune system. MHC-1 downregulation with increased PDL-1 expression of cancer cells has an important role in immune escape. MHC-1 downregulation and PDL-1 expression have been shown in many types of cancers. However, there is no study on the status of MHC-1 and PDL-1 in primary and metastatic tumor tissue. In this study, MHC-1 and PDL-1 score in primary and metastatic tumor cells was evaluated in 43 gastric cancer patients with lymph node metastasis. According to our results, the primary tumor PDL-1 score was correlated with the number of metastatic lymph nodes (r = 0.258; p = 0.024) and primary tumor size (r = 0.341; p = 0.045). A similar correlation was found between the primary tumor PDL-1 score and the metastatic tumor PDL-1 score (r = 0.213; p = 0.015). In our study, MHC-1 was found to be higher in primary tumors than metastatic tumors, although not statistically significant (p = 0.054). The results of our study showed high MHC-1 and low PDL-1 expression in primary tumors and low MHC-1 and high PDL-1 expression in metastatic tumors. These results reveal different biological characteristics of primary and metastatic tumor cells.
Collapse
|
9
|
Karakuchi N, Shimomura M, Toyota K, Hinoi T, Yamamoto H, Sadamoto S, Mandai K, Egi H, Ohdan H, Takahashi T. Spontaneous regression of transverse colon cancer with high-frequency microsatellite instability: a case report and literature review. World J Surg Oncol 2019; 17:19. [PMID: 30646898 PMCID: PMC6334436 DOI: 10.1186/s12957-018-1552-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 12/23/2018] [Indexed: 12/28/2022] Open
Abstract
Background Spontaneous regression (SR) of colorectal cancer (CRC) is extremely rare, and only few cases have been reported. Although it is not yet clarified, a plausible mechanism for SR of CRC is an immunological event. Case presentation In this report, we present the case of SR of primary CRC in a 78-year-old man. Preoperative colonoscopy was performed, and a type 2 tumor measuring 30 mm in diameter in the transverse colon was detected. The biopsy revealed a poorly differentiated adenocarcinoma. Colectomy was performed 2 months after initial colonoscopy. During the surgery, only a 10-mm ulcer harboring a polypoid lesion measuring 8.5 mm was detected in the resected tissue; no other masses or carcinoma cells were seen on histological examination. Afterwards, the biopsy specimens were reanalyzed, and immunohistological analysis verified this as adenocarcinoma with stroma-infiltrating lymphocytes. Further analysis revealed a loss of two mismatch repair proteins, suggesting sporadic high-frequency microsatellite instability (MSI-H). Conclusion According to previous literature, a common site of SR in CRC is the proximal colon, which is a feature of MSI-H CRC. However, our report showed a rare case of SR of CRC, which was in the transverse colon, with MSI-H present. This report indicates a relationship between immunological features of MSI-H and the occurrence of SR of CRC. A better understanding of this phenomenon and the mechanisms involved will have significant preventive and therapeutic implications for CRC, including anti-PD-1 immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Nozomi Karakuchi
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, 513, Jike, Saijyo-cho, Higashihiroshima, Hiroshima, 739-0041, Japan
| | - Manabu Shimomura
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, 513, Jike, Saijyo-cho, Higashihiroshima, Hiroshima, 739-0041, Japan.
| | - Kazuhiro Toyota
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, 513, Jike, Saijyo-cho, Higashihiroshima, Hiroshima, 739-0041, Japan
| | - Takao Hinoi
- Department of Surgery, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan
| | - Hideki Yamamoto
- Department of Clinical Laboratory, National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima, Japan
| | - Seiji Sadamoto
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, 513, Jike, Saijyo-cho, Higashihiroshima, Hiroshima, 739-0041, Japan
| | - Koichi Mandai
- Department of Pathology, National Hospital Organization Higashihiroshima Medical Center, Hiroshima, Japan
| | - Hiroyuki Egi
- Department of Gastrointestinal and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastrointestinal and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tadateru Takahashi
- Department of Surgery, National Hospital Organization Higashihiroshima Medical Center, 513, Jike, Saijyo-cho, Higashihiroshima, Hiroshima, 739-0041, Japan.,Department of Gastrointestinal and Transplant Surgery, Applied Life Sciences, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
10
|
Silvestris N, Brunetti O, Pinto R, Petriella D, Argentiero A, Fucci L, Tommasi S, Danza K, De Summa S. Immunological mutational signature in adenosquamous cancer of pancreas: an exploratory study of potentially therapeutic targets. Expert Opin Ther Targets 2018; 22:453-461. [PMID: 29561217 DOI: 10.1080/14728222.2018.1456530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES Adenosquamous cancer of pancreas (ASCP) is a rare variant of pancreatic adenocarcinoma (PDAC). It is characterized by poor prognosis and lacks of literature data supporting the choice of systemic therapies. The role of immunotherapy for this malignancy is still unknown. In this study, we evaluated any differences between immune-related genes of PDAC and its adenosquamous variant with the aim to characterize these histothistotypes and eventually identify potential biomarkers useful for an immune-therapy approach in ASCP. METHODS We compared the mutational status of a customized gene panel, including 41 genes involved in immunity checkpoint, inflammation and control of leukocytes, B and T cells proliferation of PDAC and ASCP. Moreover, we evaluated the immunohistochemical expression of programmed death ligand 1 (PD-L1). RESULTS We observed a status of 'hypermutation' of genes included in our panel in ASCP (22/41 mutated genes). Furtheremore, PD-L1 was found to be expressed in about 15% of the squamous component of ASCP tissue. CONCLUSION Due to genetic characteristics and to PD-L1 expression in ASCP compared to PDAC tissue, we can conclude that ASCP presents a potential sensitivity to immunological therapy.
Collapse
Affiliation(s)
- Nicola Silvestris
- a Medical Oncology Unit and Scientific Directorate , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | - Oronzo Brunetti
- b Medical Oncology Unit , Hospital of Barletta , Barletta , Italy
| | - Rosamaria Pinto
- c Pharmacogenetics and Molecular Diagnostic Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | - Daniela Petriella
- c Pharmacogenetics and Molecular Diagnostic Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | | | - Livia Fucci
- e Histopathological Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | - Stefania Tommasi
- c Pharmacogenetics and Molecular Diagnostic Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | - Katia Danza
- c Pharmacogenetics and Molecular Diagnostic Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| | - Simona De Summa
- c Pharmacogenetics and Molecular Diagnostic Unit , Istituto Tumori "Giovanni Paolo II" , Bari , Italy
| |
Collapse
|
11
|
Longo V, Gnoni A, Gardini AC, Pisconti S, Licchetta A, Scartozzi M, Memeo R, Palmieri VO, Aprile G, Santini D, Nardulli P, Silvestris N, Brunetti O. Immunotherapeutic approaches for hepatocellular carcinoma. Oncotarget 2017; 8:33897-33910. [PMID: 28420805 PMCID: PMC5464921 DOI: 10.18632/oncotarget.15406] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/01/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a cancer with a high mortality rate due to the fact that the diagnosis usually occurs at anadvanced stage. Even in case of curative surgical treatment, recurrence is common. Sorafenib and regorafenib are the only therapeutic agents that have been demonstrated to be effective in advanced HCC, thus novel curative approaches are urgently needed. Recent studies focus on the role of immune system in HCC. In fact, the unique immune response in the liver favors tolerance, which can represent a real challenge for conventional immunotherapy in these patients. Spontaneous immune responses against tumor antigens have been detected, and new immune therapies are under investigation: dendritic cell vaccination, immune-modulator strategy, and immune checkpoint inhibition. In recent years different clinical trials examining the use of immunotherapy to treat HCC have been conducted with initial promising results. This review article will summarize the literature data concerning the potential immunotherapeutic approaches in HCC patients.
Collapse
Affiliation(s)
- Vito Longo
- Medical Oncology Unit, Hospital of Taranto, Taranto, Italy
| | - Antonio Gnoni
- Medical Oncology Unit, Hospital of Gallipoli, Gallipoli, Italy
| | - Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, FC, Italy
| | | | | | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari, Monserrato, CA, Italy
| | - Riccardo Memeo
- Department of Hepatobiliary Surgery, Ospedale Regionale “F.Miulli”, Strada Pr. Acquaviva - Santeramo, Bari, Italy
| | - Vincenzo Ostilio Palmieri
- Department of Biomedical Sciences and Human Oncology, Clinica Medica “A. Murri”, University of Bari Medical School, Bari, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo Hospital ULSS 6, Vicenza, Italy
| | - Daniele Santini
- Medical Oncology Unit, University Campus Biomedico, Rome, Italy
| | - Patrizia Nardulli
- Pharmacy Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Research Centre, Istituto Tumori Giovanni Paolo II, Viale Orazio Flacco, Bari, Italy
| |
Collapse
|
12
|
Silvestris N, Brunetti O, Vasile E, Cellini F, Cataldo I, Pusceddu V, Cattaneo M, Partelli S, Scartozzi M, Aprile G, Casadei Gardini A, Morganti AG, Valentini V, Scarpa A, Falconi M, Calabrese A, Lorusso V, Reni M, Cascinu S. Multimodal treatment of resectable pancreatic ductal adenocarcinoma. Crit Rev Oncol Hematol 2017; 111:152-165. [PMID: 28259290 DOI: 10.1016/j.critrevonc.2017.01.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 01/11/2017] [Accepted: 01/24/2017] [Indexed: 01/17/2023] Open
Abstract
After a timing preoperative staging, treatment of resectable pancreatic adenocarcinoma (PDAC) includes surgery and adjuvant therapies, the former representing the initial therapeutic option and the latter aiming to reduce the incidence of both distant metastases (chemotherapy) and locoregional failures (chemoradiotherapy). Herein, we provide a critical overview on the role of multimodal treatment in PDAC and on new opportunities related to current more active poli-chemotherapy regimens, targeted therapies, and the more recent immunotherapy approaches. Moreover, an analysis of pathological markers and clinical features able to help clinicians in the selection of the best therapeutic strategy will be discussed. Lastly, the role of neoadjuvant treatment of initially resectable disease will be considered mostly in patients whose malignancy shows morphological but not clinical or biological criteria of resectability. Depending on the results of these investigational studies, today a multidisciplinary approach can offer the best address therapy for these patients.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Oronzo Brunetti
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Enrico Vasile
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.
| | - Francesco Cellini
- Radiation Oncology Department, Gemelli ART, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Ivana Cataldo
- ARC-NET Research Centre, University of Verona, Verona, Italy.
| | | | - Monica Cattaneo
- Department of Medical Oncology, University and General Hospital, Udine, Italy.
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Centre, San Raffaele Scientific Institute, 'Vita-Salute' University, Milan, Italy.
| | - Mario Scartozzi
- Medical Oncology Unit, University of Cagliari, Cagliari, Italy.
| | - Giuseppe Aprile
- Department of Medical Oncology, University and General Hospital, Udine, Italy; Department of Medical Oncology, General Hospital of Vicenza, Vicenza, Italy.
| | | | - Alessio Giuseppe Morganti
- Radiation Oncology Center, Dept. of Experimental, Diagnostic and Specialty Medicine - DIMES, University of Bologna, Italy.
| | - Vincenzo Valentini
- Radiation Oncology Department, Gemelli ART, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Aldo Scarpa
- ARC-NET Research Centre, University of Verona, Verona, Italy.
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Centre, San Raffaele Scientific Institute, 'Vita-Salute' University, Milan, Italy.
| | - Angela Calabrese
- Radiology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Vito Lorusso
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy.
| | - Michele Reni
- Medical Oncology Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Stefano Cascinu
- Modena Cancer Center, Policlinico di Modena Università di Modena e Reggio Emilia, Italy.
| |
Collapse
|
13
|
Tauriello DVF, Calon A, Lonardo E, Batlle E. Determinants of metastatic competency in colorectal cancer. Mol Oncol 2017; 11:97-119. [PMID: 28085225 PMCID: PMC5423222 DOI: 10.1002/1878-0261.12018] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/30/2016] [Accepted: 10/21/2016] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancer types and represents a major therapeutic challenge. Although initial events in colorectal carcinogenesis are relatively well characterized and treatment for early‐stage disease has significantly improved over the last decades, the mechanisms underlying metastasis – the main cause of death – remain poorly understood. Correspondingly, no effective therapy is currently available for advanced or metastatic disease. There is increasing evidence that colorectal cancer is hierarchically organized and sustained by cancer stem cells, in concert with various stromal cell types. Here, we review the interplay between cancer stem cells and their microenvironment in promoting metastasis and discuss recent insights relating to both patient prognosis and novel targeted treatment strategies. A better understanding of these topics may aid the prevention or reduction of metastatic burden.
Collapse
Affiliation(s)
- Daniele V F Tauriello
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain
| | - Alexandre Calon
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Enza Lonardo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|