1
|
Gromek P, Senkowska Z, Płuciennik E, Pasieka Z, Zhao LY, Gielecińska A, Kciuk M, Kłosiński K, Kałuzińska-Kołat Ż, Kołat D. Revisiting the standards of cancer detection and therapy alongside their comparison to modern methods. World J Methodol 2024; 14:92982. [PMID: 38983668 PMCID: PMC11229876 DOI: 10.5662/wjm.v14.i2.92982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/28/2024] [Indexed: 06/13/2024] Open
Abstract
In accordance with the World Health Organization data, cancer remains at the forefront of fatal diseases. An upward trend in cancer incidence and mortality has been observed globally, emphasizing that efforts in developing detection and treatment methods should continue. The diagnostic path typically begins with learning the medical history of a patient; this is followed by basic blood tests and imaging tests to indicate where cancer may be located to schedule a needle biopsy. Prompt initiation of diagnosis is crucial since delayed cancer detection entails higher costs of treatment and hospitalization. Thus, there is a need for novel cancer detection methods such as liquid biopsy, elastography, synthetic biosensors, fluorescence imaging, and reflectance confocal microscopy. Conventional therapeutic methods, although still common in clinical practice, pose many limitations and are unsatisfactory. Nowadays, there is a dynamic advancement of clinical research and the development of more precise and effective methods such as oncolytic virotherapy, exosome-based therapy, nanotechnology, dendritic cells, chimeric antigen receptors, immune checkpoint inhibitors, natural product-based therapy, tumor-treating fields, and photodynamic therapy. The present paper compares available data on conventional and modern methods of cancer detection and therapy to facilitate an understanding of this rapidly advancing field and its future directions. As evidenced, modern methods are not without drawbacks; there is still a need to develop new detection strategies and therapeutic approaches to improve sensitivity, specificity, safety, and efficacy. Nevertheless, an appropriate route has been taken, as confirmed by the approval of some modern methods by the Food and Drug Administration.
Collapse
Affiliation(s)
- Piotr Gromek
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zuzanna Senkowska
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Elżbieta Płuciennik
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zbigniew Pasieka
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Lin-Yong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Karol Kłosiński
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| |
Collapse
|
2
|
Ibrahimi N, Vallet L, Andre FM, Rivaletto M, Novac BM, Mir LM, Pécastaing L. An Overview of Subnanosecond Pulsed Electric Field Biological Effects: Toward Contactless Technologies for Cancer Treatment. Bioelectricity 2023. [DOI: 10.1089/bioe.2022.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Affiliation(s)
- Njomza Ibrahimi
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| | - Leslie Vallet
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Franck M. Andre
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Marc Rivaletto
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| | - Bucur M. Novac
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, United Kingdom
| | - Lluis M. Mir
- Université Paris-Saclay, CNRS, Gustave Roussy, UMR 9018, Metabolic and Systemic Aspects of Oncogenesis (METSY), Villejuif, France
| | - Laurent Pécastaing
- Laboratoire des Sciences de l'Ingénieur Appliquées à la Mécanique et au Génie Électrique–Fédération IPRA, EA4581, Université de Pau et des Pays de l'Adour/E2S UPPA, Pau, France
| |
Collapse
|
3
|
Fatima SF, Sabouni R, Garg R, Gomaa H. Recent advances in Metal-Organic Frameworks as nanocarriers for triggered release of anticancer drugs: Brief history, biomedical applications, challenges and future perspective. Colloids Surf B Biointerfaces 2023; 225:113266. [PMID: 36947901 DOI: 10.1016/j.colsurfb.2023.113266] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023]
Abstract
Metal-Organic Frameworks (MOFs) have emerged as a promising biomedical material due to its unique features such as high surface area, pore volume, variable pore size, flexible functional groups, and excellent efficiency for drug loading. In this review, we explored the use of novel and smart metal organic frameworks as drug delivery vehicles to discover a safer and more controlled mode of drug release aiming to minimize their side effects. Here, we systematically discussed the background of MOFs following a thorough review on structural and physical properties of MOFs, their synthesis techniques, and the important characteristics to establish a strong foundation for future research. Furthermore, the current status on the potential applications of MOF-based stimuli-responsive drug delivery systems, including pH-, ion-, temperature-, light-, and multiple responsive systems for the delivery of anticancer drugs has also been presented. Lastly, we discuss the prospects and challenges in implementation of MOF-based materials in the drug delivery. Therefore, this review will help researchers working in the relevant fields to enhance their understanding of MOFs for encapsulation of various drugs as well as their stimuli responsive mechanism.
Collapse
Affiliation(s)
- Syeda Fiza Fatima
- Master of Science in Biomedical Engineering Program, College of Engineering, American University of Sharjah, P.O. BOX 26666, Sharjah, United Arab Emirates
| | - Rana Sabouni
- Department of Chemical and Biological Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates.
| | - Renuka Garg
- Department of Chemical and Biological Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates
| | - Hassan Gomaa
- Department of Chemical and Biochemical Engineering, Western University, London, Canada
| |
Collapse
|
4
|
Machine learning models to prognose 30-Day Mortality in Postoperative Disseminated Cancer Patients. Surg Oncol 2022; 44:101810. [DOI: 10.1016/j.suronc.2022.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/14/2022] [Accepted: 07/03/2022] [Indexed: 11/18/2022]
|
5
|
Donlon NE, Davern M, Hayes C, Power R, Sheppard AD, Donohoe CL, Lysaght J, Reynolds JV. The immune response to major gastrointestinal cancer surgery and potential implications for adjuvant immunotherapy. Crit Rev Oncol Hematol 2022; 175:103729. [PMID: 35662586 DOI: 10.1016/j.critrevonc.2022.103729] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 12/23/2022] Open
Abstract
The perioperative period theoretically is a critical time of opportunity for the progression of pre-existing tumour micrometastasis. Therefore,the timing of introducing cancer therapies including chemotherapy, radiation therapy and immunotherapies in the postoperative period is important. A thorough exploration of the perioperative immune events at a cellular level in combination with an intricate review of available clinical rials was conducted to extrapolate the effects of oncological surgery on the perioperative immune milieu.This is timely in view of the recently published Checkmate-577 trial which demonstrated significant disease-free survival in carcinoma of the gastroesophageal junction with the use of adjuvant anti-programmed cell deathprotein 1(PD-1) immunotherapy.This review focusing in particular on perioperative immunosuppression, identifies potential modifiable factors, the effects of perioperative conditioning and optimisation, the most recent trials in the curative setting for Gastrointestinal malignancies and the new treatment avenues possible in the context of the combination of immunotherapy and major oncological gastrointestinal surgery.
Collapse
Affiliation(s)
- Noel E Donlon
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Maria Davern
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Conall Hayes
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Robert Power
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Andrew D Sheppard
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Claire L Donohoe
- Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland
| | - John V Reynolds
- Cancer Immunology and Immunotherapy Group, Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland; Department of Surgery, Trinity Translational Medicine Institute, Trinity St James's Cancer Institute Trinity College Dublin, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
6
|
Ganguli R, Franklin J, Yu X, Lin A, Heffernan DS. Machine learning methods to predict presence of residual cancer following hysterectomy. Sci Rep 2022; 12:2738. [PMID: 35177700 PMCID: PMC8854708 DOI: 10.1038/s41598-022-06585-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/24/2022] [Indexed: 12/24/2022] Open
Abstract
Surgical management for gynecologic malignancies often involves hysterectomy, often constituting the most common gynecologic surgery worldwide. Despite maximal surgical and medical care, gynecologic malignancies have a high rate of recurrence following surgery. Current machine learning models use advanced pathology data that is often inaccessible within low-resource settings and are specific to singular cancer types. There is currently a need for machine learning models to predict non-clinically evident residual disease using only clinically available health data. Here we developed and tested multiple machine learning models to assess the risk of residual disease post-hysterectomy based on clinical and operative parameters. Data from 3656 hysterectomy patients from the NSQIP dataset over 14 years were used to develop models with a training set of 2925 patients and a validation set of 731 patients. Our models revealed the top postoperative predictors of residual disease were the initial presence of gross abdominal disease on the diaphragm, disease located on the bowel mesentery, located on the bowel serosa, and disease located within the adjacent pelvis prior to resection. There were no statistically significant differences in performances of the top three models. Extreme gradient Boosting, Random Forest, and Logistic Regression models had comparable AUC ROC (0.90) and accuracy metrics (87–88%). Using these models, physicians can identify gynecologic cancer patients post-hysterectomy that may benefit from additional treatment. For patients at high risk for disease recurrence despite adequate surgical intervention, machine learning models may lay the basis for potential prospective trials with prophylactic/adjuvant therapy for non-clinically evident residual disease, particularly in under-resourced settings.
Collapse
Affiliation(s)
- Reetam Ganguli
- Brown University, Providence, USA.,Department of Surgery, Rhode Island Hospital, Brown University, Providence, USA
| | - Jordan Franklin
- Department of Computer Sciences, Georgia Institute of Technology, Atlanta, USA
| | - Xiaotian Yu
- Department of Mathematics, University of Virginia, Charlottesville, USA
| | - Alice Lin
- Warren Alpert Medical School, Providence, USA.,Department of Surgery, Rhode Island Hospital, Brown University, Providence, USA
| | - Daithi S Heffernan
- Brown University, Providence, USA. .,Warren Alpert Medical School, Providence, USA. .,Department of Surgery, Rhode Island Hospital, Brown University, Providence, USA. .,Division of Trauma/Surgical Critical Care, Division of Surgical Research, Department of Surgery, Rhode Island Hospital, Brown University, Room 207, Aldrich Building, 593 Eddy Street, Providence, RI, 02903, USA.
| |
Collapse
|
7
|
Abstract
Cancer is a multidimensional and challenging disease to handle. Current statistics reveal that we are far from satisfying cancer treatment. Taking advantage of different therapeutic agents that affect multiple pathways has been established as highly productive. Nevertheless, owing to several hindrances to conventional combination therapy, such as lack of tumor targeting, non-uniform pharmacokinetic of the combined drugs, and off-target side effects, it is well documented that this treatment approach is unlikely to address all the difficulties observed in monotherapy. Co-delivery systems could enhance the therapeutic efficacy of the combination therapy by targeting cancer cells and improving the pharmacokinetic and physicochemical properties of the therapeutic agents. Nevertheless, it seems that present knowledge in responding to the challenges in cancer treatment is still inadequate and far from optimal treatment, which highlights the urgent need for systematic studies direct to identify various aspects of co-delivery systems. Accordingly, to gather informative data, save time, and achieve superior results, the following steps are necessary: (1) implementing computational methods to predict drug-drug interactions (DDIs) in vitro and in vivo, (2) meticulous cancer studies at the cellular and molecular levels to obtain specific criteria for selecting preclinical and clinical models, (3) extensive physiological and pharmacokinetic study of nanocarriers behavior in preclinical models, and (4) finding the optimal formulation and analyzing its behavior in cellular and animal models facilitates bridging in vivo models to clinical trials. This review aims to deliver an overview of co-delivery systems, rationales, and suggestions for further studies in this field.
Collapse
|
8
|
He G, Yan X, Miao Z, Qian H, Ma Y, Xu Y, Gao L, Lu Y, Zha Z. Anti-inflammatory catecholic chitosan hydrogel for rapid surgical trauma healing and subsequent prevention of tumor recurrence. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.02.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
9
|
Redmond HP, Neary PM, Jinih M, O'Connell E, Foley N, Pfirrmann RW, Wang JH, O'Leary DP. RandomiSed clinical trial assessing Use of an anti-inflammatoRy aGent in attenUating peri-operatiVe inflAmmatioN in non-meTastatic colon cancer - the S.U.R.G.U.V.A.N.T. trial. BMC Cancer 2018; 18:794. [PMID: 30081854 PMCID: PMC6091184 DOI: 10.1186/s12885-018-4641-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/27/2018] [Indexed: 12/22/2022] Open
Abstract
Background Peri-operative inflammation has been extensively highlighted in cancer patients as detrimental. Treatment strategies to improve survival for cancer patients through targeting peri-operative inflammation have yet to be devised. Methods We conducted a multi-centre, randomised controlled clinical trial using Taurolidine in non-metastatic colon cancer patients. Patients were randomly assigned to receive Taurolidine or a placebo. The primary endpoint for the study was the mean difference in day 1 IL-6 levels. Secondary clinical endpoints included rates of post-operative infections and tumor recurrence. Results A total of 293 patients were screened for trial inclusion. Sixty patients were randomised. Twenty-eight patients were randomised to placebo and 32 patients to Taurolidine. IL-6 levels were equivalent on day 1 post-operatively in both groups. However, IL-6 levels were significantly attenuated over the 7 day study period in the Taurolidine group compared to placebo (p = 0.04). In addition, IL-6 levels were significantly lower at day 7 in the Taurolidine group (p = 0.04). There were 2 recurrences in the placebo group at 2 years and 1 in the Taurolidine group. The median time to recurrence was 19 months in the Placebo group and 38 months in the Taurolidine group (p = 0.27). Surgical site infection was reduced in the Taurolidine treated group (p = 0.09). Conclusion Peri-operative use of Taurolidine significantly attenuated circulating IL-6 levels in the initial 7 day post-operative period in a safe manner. Future studies are required to establish the impact of IL-6 attenuation on survival outcomes in colon cancer. Trial registration The trial was registered with EudraCT (year = 2008, registration number = 005570–12) and ISRCTN (year = 2008, registration number = 77,829,558).
Collapse
Affiliation(s)
- H Paul Redmond
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Peter M Neary
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Marcel Jinih
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Emer O'Connell
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Niamh Foley
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Rolf W Pfirrmann
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - Jiang H Wang
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland
| | - D Peter O'Leary
- Surguvant Research Centre, Cork University Hospital, Cork, Ireland.
| |
Collapse
|
10
|
Grewal S, Korthouwer R, Bögels M, Braster R, Heemskerk N, Budding AE, Pouw SM, van Horssen J, Ankersmit M, Meijerink J, van den Tol P, Oosterling S, Bonjer J, Gül N, van Egmond M. Spillage of bacterial products during colon surgery increases the risk of liver metastases development in a rat colon carcinoma model. Oncoimmunology 2018; 7:e1461302. [PMID: 30228930 PMCID: PMC6140552 DOI: 10.1080/2162402x.2018.1461302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/28/2018] [Accepted: 03/30/2018] [Indexed: 01/03/2023] Open
Abstract
Surgical resection of the primary tumor provides the best chance of cure for patients with colorectal carcinoma (CRC). However, bacterial translocation during intestinal surgery has been correlated with poor long-term oncological outcome. Therefore, we investigated the influence of bacterial contamination during colon surgery on CRC liver metastases development. Blood and liver samples of patients undergoing resection of primary CRC or liver metastases were collected. Cell numbers, activation markers and inflammatory mediators were determined. Tumor cell adhesion and outgrowth after sham- or colectomy operations were determined in a rat model, in which tumor cells had been injected into the portal vein. White blood cells and granulocytes were increased in per- and post-operative patient blood samples. IL-6 was also increased post-operatively compared to the preoperative level. Expression of NOX-2, NOX-4 and polymorphonuclear cells (PMNs) numbers were elevated in post-operative human liver samples. In vitro stimulation of macrophages with plasma of rats after colectomy resulted in production of reactive oxygen species (ROS). Colectomy in rats increased D-lactate levels in plasma, supporting bacterial translocation. Decreased expression of tight junction molecules and increased tumor cell adhesion and outgrowth was observed. Treatment with a selective decontamination of the digestive tract (SDD) cocktail decreased tumor cell adherence after colectomy. In conclusion, postoperative bacterial translocation may activate liver macrophages and PMNs, resulting in ROS production. As we previously showed that ROS release led to liver vasculature damage, circulating tumor cells may adhere to exposed extracellular matrix and grow out into liver metastases. This knowledge is pivotal for development of therapeutic strategies to prevent surgery-induced liver metastases development.
Collapse
Affiliation(s)
- Simran Grewal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands.,Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Rianne Korthouwer
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Marijn Bögels
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands.,Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Rens Braster
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Niels Heemskerk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Andries E Budding
- Department of Medical Microbiology and Infection Control, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Stephan M Pouw
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Jack van Horssen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Marjolein Ankersmit
- Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Jeroen Meijerink
- Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Petrousjka van den Tol
- Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Steven Oosterling
- Department of Surgery, Spaarne Gasthuis, PO Box 417, 2000 AK Haarlem, the Netherlands
| | - Jaap Bonjer
- Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| | - Nuray Gül
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, PO Box 7057, 1007 MB Amsterdam, the Netherlands.,Department of Surgery, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, the Netherlands
| |
Collapse
|
11
|
Tohme S, Simmons RL, Tsung A. Surgery for Cancer: A Trigger for Metastases. Cancer Res 2017; 77:1548-1552. [PMID: 28330928 DOI: 10.1158/0008-5472.can-16-1536] [Citation(s) in RCA: 408] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/03/2016] [Accepted: 12/07/2016] [Indexed: 12/12/2022]
Abstract
Surgery is a crucial intervention and provides a chance of cure for patients with cancer. The perioperative period is characterized by an increased risk for accelerated growth of micrometastatic disease and increased formation of new metastatic foci. The true impact for cancer patients remains unclear. This review summarizes the often fragmentary clinical and experimental evidence supporting the role of surgery and inflammation as potential triggers for disease recurrence. Surgery induces increased shedding of cancer cells into the circulation, suppresses antitumor immunity allowing circulating cells to survive, upregulates adhesion molecules in target organs, recruits immune cells capable of entrapping tumor cells, and induces changes in the target tissue and in the cancer cells themselves to enhance migration and invasion to establish at the target site. Surgical trauma induces local and systemic inflammatory responses that can also contribute to the accelerated growth of residual and micrometastatic disease. Furthermore, we address the role of perioperative factors, including anesthesia, transfusions, hypothermia, and postoperative complications, as probable deleterious factors contributing to early recurrence. Through the admittedly limited understanding of these processes, we will attempt to provide suggestions for potential new therapeutic approaches to target the protumorigenic perioperative window and ultimately improve long-term oncological outcomes. Cancer Res; 77(7); 1548-52. ©2017 AACR.
Collapse
Affiliation(s)
- Samer Tohme
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Richard L Simmons
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
12
|
Glatz T, Lederer AK, Kulemann B, Seifert G, Holzner PA, Hopt UT, Hoeppner J, Marjanovic G. The degree of local inflammatory response after colonic resection depends on the surgical approach: an observational study in 61 patients. BMC Surg 2015; 15:108. [PMID: 26444274 PMCID: PMC4596306 DOI: 10.1186/s12893-015-0097-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 10/01/2015] [Indexed: 01/04/2023] Open
Abstract
Background Clinical data indicate that laparoscopic surgery reduces postoperative inflammatory response and benefits patient recovery. Little is known about the mechanisms involved in reduced systemic and local inflammation and the contribution of reduced trauma to the abdominal wall and the parietal peritoneum. Methods Included were 61 patients, who underwent elective colorectal resection without intraabdominal complications; 17 received a completely laparoscopic, 13 a laparoscopically- assisted procedure and 31 open surgery. Local inflammatory response was quantified by measurement of intraperitoneal leukocytes and IL-6 levels during the first 4 days after surgery. Results There was no statistical difference between the groups in systemic inflammatory parameters and intraperitoneal leukocytes. Intraperitoneal interleukin-6 was significantly lower in the laparoscopic group than in the laparoscopically-assisted and open group on postoperative day 1 (26.16 versus 43.25 versus 40.83 ng/ml; p = 0.001). No difference between the groups was recorded on POD 2–4. Intraperitoneal interleukin-6 showed a correlation with duration of hospital stay on POD 1 (0.233, p = 0.036), but not on POD 2–4. Patients who developed a surgical wound infection showed higher levels of intraperitoneal interleukin-6 on postoperative day 2–4 (POD 2: 42.56 versus 30.02 ng/ml, p = 0.03), POD 3: 36.52 versus 23.62 ng/ml, p = 0.06 and POD 4: 34.43 versus 19.99 ng/ml, p = 0.046). Extraabdominal infections had no impact. Conclusion The analysis shows an attenuated intraperitoneal inflammatory response on POD 1 in completely laparoscopically-operated patients, associated with a quicker recovery. This effect cannot be observed in patients, who underwent a laparoscopically-assisted or open procedure. Factors inflicting additional trauma to the abdominal wall and parietal peritoneum promote the intraperitoneal inflammation process.
Collapse
Affiliation(s)
- Torben Glatz
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany.
| | - Ann-Kathrin Lederer
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Birte Kulemann
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Gabriel Seifert
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Philipp Anton Holzner
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Ulrich Theodor Hopt
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Jens Hoeppner
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| | - Goran Marjanovic
- Department of General and Visceral Surgery, Albert Ludwigs University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
13
|
Tai LH, Auer R. Attacking Postoperative Metastases using Perioperative Oncolytic Viruses and Viral Vaccines. Front Oncol 2014; 4:217. [PMID: 25161958 PMCID: PMC4130104 DOI: 10.3389/fonc.2014.00217] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/30/2014] [Indexed: 01/01/2023] Open
Abstract
Surgical resection of solid primary malignancies is a mainstay of therapy for cancer patients. Despite being the most effective treatment for these tumors, cancer surgery has been associated with impaired metastatic clearance due to immunosuppression. In preclinical surgery models and human cancer patients, we and others have demonstrated a profound suppression of both natural killer (NK) and T cell function in the postoperative period and this plays a major role in the enhanced development of metastases following surgery. Oncolytic viruses (OV) were originally designed to selectively infect and replicate in tumors, with the primary objective of directly lysing cancer cells. It is becoming increasingly clear, however, that OV infection results in a profound inflammatory reaction within the tumor, initiating innate and adaptive immune responses against it that is critical for its therapeutic benefit. This anti-tumor immunity appears to be mediated predominantly by NK and cytotoxic T cells. In preclinical models, we found that preoperative OV prevents postoperative NK cell dysfunction and attenuates tumor dissemination. Due to theoretical safety concerns of administering live virus prior to surgery in cancer patients, we characterized safe, attenuated versions of OV, and viral vaccines that could stimulate NK cells and reduce metastases when administered in the perioperative period. In cancer patients, we observed that in vivo infusion with oncolytic vaccinia virus and ex vivo stimulation with viral vaccines promote NK cell activation. These preclinical studies provide a novel and clinically relevant setting for OV therapy. Our challenge is to identify safe and promising OV therapies that will activate NK and T cells in the perioperative period preventing the establishment of micrometastatic disease in cancer patients.
Collapse
Affiliation(s)
- Lee-Hwa Tai
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Rebecca Auer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
14
|
Kahokehr AA. Intraperitoneal wound in abdominal surgery. World J Crit Care Med 2013; 2:1-3. [PMID: 24701409 PMCID: PMC3953863 DOI: 10.5492/wjccm.v2.i1.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 07/14/2012] [Accepted: 12/05/2012] [Indexed: 02/06/2023] Open
Abstract
The intraperitoneal wound is often forgotten after transperitoneal surgery. This review is a on the peritoneum and the implications of peritoneal injury after surgery. This review will focus on the intraperitoneal wound response after surgical injury.
Collapse
Affiliation(s)
- Arman Adam Kahokehr
- Arman Adam Kahokehr, Department of Surgery, University of Auckland, Auckland 1021, New Zealand
| |
Collapse
|
15
|
A prospective case-control study of the local and systemic cytokine response after laparoscopic versus open colonic surgery. J Surg Res 2010; 173:278-85. [PMID: 21195431 DOI: 10.1016/j.jss.2010.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 09/11/2010] [Accepted: 10/13/2010] [Indexed: 12/23/2022]
Abstract
BACKGROUND There is a sequential, high concentration cytokine response after major abdominal surgery. The magnitude of this response has been directly linked to postoperative metabolic derangement, ileus, adhesions, and oncological outcomes. We aimed to compare the local and systemic cytokine response in laparoscopic and open colonic surgery and relate this to postoperative recovery parameters. METHODS Using a prospectively collected patient database, we compared a Study Group (n = 50) of patients undergoing elective laparoscopic colonic resection with a Control Group (n = 25) of patients undergoing equivalent open colonic surgery within an ERAS program. Patients were matched for age, gender, BMI, ASA, Cr Possum, side of resection, diagnosis, and histologic stage. Plasma and peritoneal fluid concentrations of IL-6, IL-8, IL-10, and TNFα were measured at 20-24 h after surgery. The Surgical Recovery Score was determined pre-operatively and at 3, 7, 30, and 60 d postoperatively. All data were prospectively collected, and a priori definitions were used for discharge parameters, complications, and complication severity. RESULTS Peritoneal fluid IL-6 concentration was lower after laparoscopic surgery. There were no significant differences in the other cytokines measured, or in any postoperative recovery outcomes. Significant correlations were found between cytokine levels and discharge criteria achievement, day stay, postoperative complications, and the Surgical Recovery Score. CONCLUSION With the exception of a lower peritoneal IL-6 level, the systemic and peritoneal cytokine response at 20-24 h is similar after laparoscopic versus open colonic resection within an ERAS program, with corresponding equivalent rates of postoperative recovery.
Collapse
|
16
|
Peritoneal damage: the inflammatory response and clinical implications of the neuro-immuno-humoral axis. World J Surg 2010; 34:704-20. [PMID: 20049432 DOI: 10.1007/s00268-009-0382-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND The peritoneum is a bilayer serous membrane that lines the abdominal cavity. We present a review of peritoneal structure and physiology, with a focus on the peritoneal inflammatory response to surgical injury and its clinical implications. METHODS We conducted a nonsystematic clinical review. A search of the Ovid MEDLINE database from 1950 through January 2009 was performed using the following search terms: peritoneum, adhesions, cytokine, inflammation, and surgery. RESULTS The peritoneum is a metabolically active organ, responding to insult through a complex array of immunologic and inflammatory cascades. This response increases with the duration and extent of injury and is central to the concept of surgical stress, manifesting via a combination of systemic effects, and local neural pathways via the neuro-immuno-humoral axis. There may be a decreased systemic inflammatory response after minimally invasive surgery; however, it is unclear whether this is due to a reduced local peritoneal reaction. CONCLUSIONS Interventions that dampen the peritoneal response and/or block the neuro-immuno-humoral pathway should be further investigated as possible avenues of enhancing recovery after surgery, and reducing postoperative complications.
Collapse
|
17
|
van der Bij GJ, Oosterling SJ, Beelen RHJ, Meijer S, Coffey JC, van Egmond M. The perioperative period is an underutilized window of therapeutic opportunity in patients with colorectal cancer. Ann Surg 2009; 249:727-34. [PMID: 19387333 DOI: 10.1097/sla.0b013e3181a3ddbd] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE In this review, we address the underlying mechanisms by which surgery augments metastases outgrowth and how these insights can be used to develop perioperative therapeutic strategies for prevention of tumor recurrence. SUMMARY BACKGROUND DATA Surgical removal of the primary tumor provides the best chance of long-term disease-free survival for patients with colorectal cancer (CRC). Unfortunately, a significant part of CRC patients will develop metastases, even after successful resection of the primary tumor. Paradoxically, it is now becoming clear that surgery itself contributes to development of both local recurrences and distant metastases. METHODS Data for this review were identified by searches of PubMed and references from relevant articles using the search terms "surgery," "CRC," and "metastases." RESULTS Surgical trauma and concomitant wound-healing processes induce local and systemic changes, including impairment of tissue integrity and production of inflammatory mediators and angiogenic factors. This can lead to immune suppression and enhanced growth or adhesion of tumor cells, all of which increase the chance of exfoliated tumor cells developing into secondary malignancies. CONCLUSIONS Because surgery remains the appropriate and necessary means of treatment for most CRC patients, new adjuvant therapeutic strategies that prevent tumor recurrence after surgery need to be explored since the perioperative therapeutic window of opportunity offers promising means of improving patient outcome but is unfortunately underutilized.
Collapse
Affiliation(s)
- Gerben J van der Bij
- Department of Surgical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
18
|
Shantha Kumara HMC, Kirman I, Feingold D, Cekic V, Nasar A, Arnell T, Balik E, Hoffman A, Baxter R, Conte S, Whelan RL. Perioperative GMCSF limits the proangiogenic plasma protein changes associated with colorectal cancer resection. Eur J Surg Oncol 2008; 35:295-301. [PMID: 18782657 DOI: 10.1016/j.ejso.2008.07.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 05/20/2008] [Accepted: 07/21/2008] [Indexed: 01/04/2023] Open
Abstract
AIMS Colorectal resection (CR) increases plasma VEGF levels which may promote residual tumor growth. This study assessed the effect of perioperative GMCSF on plasma levels of sVEGFR1, Ang-1 and Ang-2 and also the impact of post-GMCSF plasma on in vitro endothelial cell (EC) growth and invasion. Ang-2 increases while sVEGFR1 and Ang-1 impede angiogenesis. METHODS Fifty-nine CR cancer patients were randomized to 7 perioperative doses of GMCSF or saline for 3days prior and 4days after CR. Blood samples were taken pre-drug (PreRx) and on several postoperative days (POD). Protein levels were assessed and PreRx and POD 5 plasma added to EC cultures after which branch point formation (ECBPF) and invasion (ECI) were measured. RESULTS sVEGFR1 levels were significantly higher on POD 1 and POD 5 in both groups but the GMCSF POD 5 level was twice the control value (p=0.002). Ang-2 levels were higher on PODs 1 and 5 in both groups (p<0.05) but the control POD 5 value (vs. GMCSF) was greater (p=0.03). Ang-1 decreases were noted in all (p=not significant, ns). The control group POD 5 ECBPF was 35.8% greater than Pre Rx (p=0.001) while the GMCSF result was 18.0% lower (p=ns); the control POD 5 median percent change from baseline was greater than the GMCSF result(p=0.008). The POD 5 ECI was +12.2% for the control group vs. baseline (p=ns) and -17.2% for the GMCSF group (p=ns): the control median percent change was greater than in the GMCSF group(p=0.045). CONCLUSION CR-related plasma changes are proangiogenic (>Ang-2) and anti-angiogenic (>sVEGFR1); the net effect is promotion of in vitro ECBPF. GMCSF limits the proangiogenic changes (higher POD 5 sVEGFR1 levels and lower Ang-2 elevations, lower POD 5 ECBPF and ECI). The clinical import of these effects is unclear; perioperative GMCSF has anti-angiogenic plasma effects that may limit tumor growth. Further investigation is warranted.
Collapse
Affiliation(s)
- H M C Shantha Kumara
- Department of Surgery, Columbia University, New York Presbyterian Hospital, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Liu Z, Jiang M, Yan F, Xu L, Zhao J, Ju H. Multipoint quantification of multimarker genes in peripheral blood and micrometastasis characteristic in peri-operative esophageal cancer patients. Cancer Lett 2008; 261:46-54. [DOI: 10.1016/j.canlet.2007.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2007] [Revised: 11/01/2007] [Accepted: 11/05/2007] [Indexed: 10/22/2022]
|