1
|
Yang W, Qian C, Luo J, Chen C, Feng Y, Dai N, Li X, Xiao H, Yang Y, Li M, Li C, Wang D. First in human intraarterial delivery of tislelizumab for the treatment of pMMR locally advanced rectal cancer: A single-arm, open label, phase II clinical trial. Transl Oncol 2024; 50:102154. [PMID: 39405605 DOI: 10.1016/j.tranon.2024.102154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/03/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Intravenous immune checkpoint inhibitors (ICIs) have shown efficacy in treating locally advanced rectal cancer (LARC), but concerns about systemic toxicity persist. This study developed a unique approach termed chemo-immuno-embolization with transcatheter rectal arterial intervention (CIETAI), aiming to enhance the anti-tumor response while minimizing systemic toxicity. METHOD This is a prospective, single-arm, phase II clinical trial conducted in Daping hospital. Patients with previously untreated stage II/III LARC underwent preoperative CIETAI combined with PD-1 inhibitor tislelizumab plus oxaliplatin, followed by standard concomitant chemoradiotherapy (capecitabine and 50.4 Gy radiation). Intravenous tislelizumab was administered for an additional two cycles. RESULTS Between January 2023 and December 2023, a total of 38 patients were enrolled. As the primary endpoint, 17 (44.74 %) patients achieved pathological complete response (TRG0), with a major pathologic response (MPR) rate of 65.79 %. The anal preservation rate was 84.21 % (32/38), and importantly, 15 of 21 patients with low rectal cancer achieved organ preservation with functional maintenance. Eight patients experienced grade 3-4 adverse events (AEs). All immune-related AEs were grade 1-2, with the most common being endocrine toxicity (5/6, 83.33 %). No grade 5 AEs occurred. CONCLUSION This study provides preliminary evidence supporting the safety and efficacy of intraarterial tislelizumab delivery in the neoadjuvant setting for LARC. These promising results encourage further exploration in larger cohorts to validate the clinical impact of this novel CIETAI strategy. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05957016.
Collapse
Affiliation(s)
- Weina Yang
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Chengyuan Qian
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing University, Chongqing 402260, China
| | - Jiamin Luo
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Chuan Chen
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Yan Feng
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Nan Dai
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Xuemei Li
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - He Xiao
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Yuxin Yang
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Mengxia Li
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China
| | - Chunxue Li
- Department of General Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China.
| | - Dong Wang
- Cancer Center, Daping Hospital, Army Medical University, No.10 Changjiang Zhi Road, Daping Yuzhong District, Chongqing 400042, China; Department of Oncology, Chongqing University Qianjiang Hospital, Chongqing University, Chongqing, 409099, China.
| |
Collapse
|
2
|
Yang G, Cao Y, Yang X, Cui T, Tan NZV, Lim YK, Fu Y, Cao X, Bhandari A, Enikeev M, Efetov S, Balaban V, He M. Advancements in nanomedicine: Precision delivery strategies for male pelvic malignancies - Spotlight on prostate and colorectal cancer. Exp Mol Pathol 2024; 137:104904. [PMID: 38788248 DOI: 10.1016/j.yexmp.2024.104904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Pelvic malignancies consistently pose significant global health challenges, adversely affecting the well-being of the male population. It is anticipated that clinicians will continue to confront these cancers in their practice. Nanomedicine offers promising strategies that revolutionize the treatment of male pelvic malignancies by providing precise delivery methods that aim to improve the efficacy of therapeutic outcomes while minimizing side effects. Nanoparticles are designed to encapsulate therapeutic agents and selectively target cancer cells. They can also be loaded with theragnostic agents, enabling multifunctional capabilities. OBJECTIVE This review aims to summarize the latest nanomedicine research into clinical applications, focusing on nanotechnology-based treatment strategies for male pelvic malignancies, encompassing chemotherapy, radiotherapy, immunotherapy, and other cutting-edge therapies. The review is structured to assist physicians, particularly those with limited knowledge of biochemistry and bioengineering, in comprehending the functionalities and applications of nanomaterials. METHODS Multiple databases, including PubMed, the National Library of Medicine, and Embase, were utilized to locate and review recently published articles on advancements in nano-drug delivery for prostate and colorectal cancers. CONCLUSION Nanomedicine possesses considerable potential in improving therapeutic outcomes and reducing adverse effects for male pelvic malignancies. Through precision delivery methods, this emerging field presents innovative treatment modalities to address these challenging diseases. Nevertheless, the majority of current studies are in the preclinical phase, with a lack of sufficient evidence to fully understand the precise mechanisms of action, absence of comprehensive pharmacotoxicity profiles, and uncertainty surrounding long-term consequences.
Collapse
Affiliation(s)
- Guodong Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Te Cui
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yuen Kai Lim
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Yu Fu
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xinren Cao
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Aanchal Bhandari
- HBT Medical College and Dr. R N Cooper Municipal General Hospital, Mumbai, India
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia
| | - Sergey Efetov
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vladimir Balaban
- Clinic of Coloproctology and Minimally Invasive Surgery, Sechenov University, Moscow, Russia
| | - Mingze He
- Institute for Urology and Reproductive Health, Sechenov University, Moscow, Russia.
| |
Collapse
|
3
|
Nguyen NP, Mohammadianpanah M, SunMyint A, Page BR, Vinh-Hung V, Gorobets O, Arenas M, Mazibuko T, Giap H, Vasileiou M, Dutheil F, Tuscano C, Karlsson ULFL, Dahbi Z, Natoli E, Li E, Kim L, Oboite J, Oboite E, Bose S, Vuong T. Immunotherapy and radiotherapy for older patients with locally advanced rectal cancer unfit for surgery or decline surgery: a practical proposal by the International Geriatric Radiotherapy Group. Front Oncol 2024; 14:1325610. [PMID: 38463223 PMCID: PMC10921228 DOI: 10.3389/fonc.2024.1325610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/16/2024] [Indexed: 03/12/2024] Open
Abstract
The standard of care for locally advanced rectal cancer is total neoadjuvant therapy followed by surgical resection. Current evidence suggests that selected patients may be able to delay or avoid surgery without affecting survival rates if they achieve a complete clinical response (CCR). However, for older cancer patients who are too frail for surgery or decline the surgical procedure, local recurrence may lead to a deterioration of patient quality of life. Thus, for clinicians, a treatment algorithm which is well tolerated and may improve CCR in older and frail patients with rectal cancer may improve the potential for prolonged remission and potential cure. Recently, immunotherapy with check point inhibitors (CPI) is a promising treatment in selected patients with high expression of program death ligands receptor 1 (PD- L1). Radiotherapy may enhance PD-L1 expression in rectal cancer and may improve response rate to immunotherapy. We propose an algorithm combining immunotherapy and radiotherapy for older patients with locally advanced rectal cancer who are too frail for surgery or who decline surgery.
Collapse
Affiliation(s)
- Nam P. Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Mohammad Mohammadianpanah
- Colorectal Research Center, Department of Radiation Oncology, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arthur SunMyint
- Department of Radiation Oncology, Clatterbridge Cancer Center, Liverpool, United Kingdom
| | - Brandi R. Page
- Department of Radiation Oncology, Johns Hopkins University, Bethesda, MD, United States
| | - Vincent Vinh-Hung
- Department of Radiation Oncology, Institut Bergonie, Bordeaux, France
| | - Olena Gorobets
- Department of Oral Surgery, Martinique University, Fort de France, France
| | - Meritxell Arenas
- Department of Radiation Oncology, Sant Joan de Reus University, University of Rovira, I Virgili, Tarragona, Spain
| | - Thandeka Mazibuko
- Department of Radiation Oncology, International Geriatric Radiotherapy Group, Washington, DC, United States
| | - Huan Giap
- Department of Radiation Oncology, Medical University of South Carolina, Charleston, SC, United States
| | - Maria Vasileiou
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Fabien Dutheil
- Department of Radiation Oncology, Clinique Sainte Clotilde, Saint Denis, La Reunion, Saint Denis, France
| | - Carmelo Tuscano
- Department of Radiation Oncology, A.O Bianchi Melacrino, Reggio Calabria, Italy
| | - ULF Lennart Karlsson
- Department of Radiation Oncology, International Geriatric Radiotherapy Group, Washington, DC, United States
| | - Zineb Dahbi
- Department of Radiation Oncology, Mohammed VI University of Health Sciences, Casablanca, Morocco
| | - Elena Natoli
- Department of Radiation Oncology, University of Bologna, Bologna, Italy
| | - Eric Li
- Department of Pathology, Howard University, Washington, DC, United States
| | - Lyndon Kim
- Division of Neurooncology, Mt Sinai Hospital, New York, NY, United States
| | - Joan Oboite
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Eromosele Oboite
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Satya Bose
- Department of Radiation Oncology, Howard University, Washington, DC, United States
| | - Te Vuong
- Department of Radiation Oncology, Mc Gill University, Montreal, Canada
| |
Collapse
|
4
|
Malik A, Bhatia JK, Sahai K, Boruah D, Sharma A. Evaluating morphological features for predicting microsatellite instability status in colorectal cancer. Med J Armed Forces India 2022; 78:S96-S104. [PMID: 36147411 PMCID: PMC9485851 DOI: 10.1016/j.mjafi.2021.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/28/2021] [Indexed: 10/20/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the commonest cancers worldwide, with incidence rates in India being around 4%. It is a heterogeneous disease with multiple established prognostic factors. Ten to fifteen percent originate from microsatellite instability (MSI) pathway, characterized by defect in mismatch repair (MMR) gene. Identification of MMR defective protein is relevant for diagnosis, prognosis, and prediction. Certain clinical and histological features are known to be associated with defective MMR genes. The objectives of this study are to find the prevalence of MSI in CRC to identify features associated with MSI and assess the value of histopathology in predicting MSI. Methods We evaluated various clinical and histological parameters for identifying prognostically favorable colon cancers in a tertiary hospital. One hundred fifty colon cancers were evaluated, and MSI status was correlated with clinicopathologic variables. Results The prevalence of MSI in CRC was found to be 11.3%. The factors associated with MSI were tumor differentiation, stage, tumor site, tumor size, tumor-infiltrating lymphocytes, Crohn's-like lymphoid reaction, and dirty necrosis. We have defined a "P" score for prediction of MSI using the clinicohistological parameters, which could be used to select patients who are to be tested for MSI. Conclusion Assessment of clinical and histopathological features will help in patient stratification and selection of patients for MSI testing. The evaluation is economical, reproducible, and easy to apply.
Collapse
Affiliation(s)
| | - Jasvinder Kaur Bhatia
- Senior Adviser & Head (Pathology), Command Hospital (Eastern Command) Kolkata, India
| | | | - Dibyajyoti Boruah
- Scientist ‘E’, Department of Pathology, Armed Forces Medical College, Pune, India
| | - A. Sharma
- Classified Specialist (Pathology), INHS Kalyani, Visakhapatnam, India
| |
Collapse
|
5
|
Shi M, Chen Y, Ji D. The implications from the interplay of neoadjuvant chemoradiotherapy and the immune microenvironment in rectal cancer. Future Oncol 2022; 18:3229-3244. [PMID: 36017694 DOI: 10.2217/fon-2022-0061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neoadjuvant chemoradiotherapy (nCRT) is recommended for the treatment of locally advanced rectal cancer. Even though the combination of nCRT and immune checkpoint inhibitors (ICIs) has received much attention, the specific combination modes and dose fractions in radiotherapy (RT) are still indistinct. This review focuses on the immunological mechanism involved in nCRT, the clinical efficacy, the immunological effect of different combined strategies, concurrent or sequential nCRT plus ICIs, long-course RT and short-course RT. This review discusses the impact of nCRT on tumor immunity and summarizes the availability of different dose fractions in RT and distinct combined strategies, aiming at providing clues for optimal neoadjuvant therapy options that maximize efficacy and minimize side effects.
Collapse
Affiliation(s)
- Mengyuan Shi
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| | - Yongkang Chen
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| | - Dengbo Ji
- Key laboratory of Carcinogenesis & Translational Research (Ministry of Education), Department of Gastrointestinal Surgery III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd., Haidian District, Beijing, 100142, China
| |
Collapse
|
6
|
Sclafani F, Corrò C, Koessler T. Debating Pros and Cons of Total Neoadjuvant Therapy in Rectal Cancer. Cancers (Basel) 2021; 13:cancers13246361. [PMID: 34944980 PMCID: PMC8699289 DOI: 10.3390/cancers13246361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Rectal cancers represent one third of all colorectal tumours. Patients diagnosed with localised colon cancer undergo surgery upfront, likely followed by adjuvant chemotherapy. Those diagnosed with localised rectal cancer, however, frequently benefit from neoadjuvant treatments with either radiotherapy or chemoradiotherapy before undergoing surgery. On the other hand, the benefit of adjuvant chemotherapy in this setting is more controversial. The main challenges in treating patients affected by rectal cancer encompass: decreasing the risks of local relapse and distant metastases, preserving the sphincter and minimising treatment-associated functional sequelae, and improving overall survival. Some of these fuelled the concept of total neoadjuvant therapy, namely giving all available treatments including radiotherapy and systemic chemotherapy before surgery. Here, we critically review the pros and cons of such a treatment strategy, but also discuss the biological rational to support neoadjuvant treatment intensification. Abstract Recently, two large, randomised phase III clinical trials of total neoadjuvant therapy (TNT) in locally advanced rectal cancer were published (RAPIDO and PRODIGE 23). These two trials compared short-course radiotherapy (SCRT) followed by chemotherapy with standard chemoradiotherapy (CRT) and chemotherapy followed by CRT with standard CRT, respectively. They showed improvement in some of the outcomes such as distant recurrence and pathological complete response (pCR). No improvement, however, was observed in local disease control or the de-escalation of surgical procedures. Although it seems lawful to integrate TNT within the treatment algorithm of localised stage II and III rectal cancer, many questions remain unanswered, including which are the optimal criteria to identify patients who are most likely to benefit from this intensive treatment. Instead of providing a sterile summary of trial results, we put these in perspective in a pros and cons manner. Moreover, we discuss some biological aspects of rectal cancer, which may provide some insights into the current decision-making process, and represent the basis for the future development of alternative, more effective treatment strategies.
Collapse
Affiliation(s)
- Francesco Sclafani
- Department of Medical Oncology, Institut Jules Bordet, Rue Meylemeersch 90, 1070 Anderlecht, Belgium;
- Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070 Brussels, Belgium
| | - Claudia Corrò
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland;
- Swiss Cancer Center Léman, Geneva and Lausanne, 1005 Lausanne, Switzerland
| | - Thibaud Koessler
- Translational Research Center in Onco-Hematology, Department of Medicine, Faculty of Medicine, University of Geneva, 1205 Geneva, Switzerland;
- Swiss Cancer Center Léman, Geneva and Lausanne, 1005 Lausanne, Switzerland
- Department of Oncology, Geneva University Hospital, 1205 Geneva, Switzerland
- Correspondence:
| |
Collapse
|
7
|
Zaborowski AM, Winter DC, Lynch L. The therapeutic and prognostic implications of immunobiology in colorectal cancer: a review. Br J Cancer 2021; 125:1341-1349. [PMID: 34302062 PMCID: PMC8575924 DOI: 10.1038/s41416-021-01475-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/13/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer represents the second leading cause of cancer-related death worldwide. The therapeutic field of immuno-oncology has rapidly gained momentum, with strikingly promising results observed in clinical practice. Increasing emphasis has been placed on the role of the immune response in tumorigenesis, therapy and predicting prognosis. Enhanced understanding of the dynamic and complex tumour-immune microenvironment has enabled the development of molecularly directed, individualised treatment. Analysis of intra-tumoural lymphocyte infiltration and the dichotomisation of colorectal cancer into microsatellite stable and unstable disease has important therapeutic and prognostic implications, with potential to capitalise further on this data. This review discusses the latest evidence surrounding the tumour biology and immune landscape of colorectal cancer, novel immunotherapies and the interaction of the immune system with each apex of the tripartite of cancer management (oncotherapeutics, radiotherapy and surgery). By utilising the synergy of chemotherapeutic agents and immunotherapies, and identifying prognostic and predictive immunological biomarkers, we may enter an era of unprecedented disease control, survivorship and cure rates.
Collapse
Affiliation(s)
- Alexandra M. Zaborowski
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Des C. Winter
- grid.412751.40000 0001 0315 8143Centre for Colorectal Disease, St. Vincent’s University Hospital, Dublin 4, Ireland ,grid.7886.10000 0001 0768 2743School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Lydia Lynch
- grid.8217.c0000 0004 1936 9705School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland ,grid.38142.3c000000041936754XHarvard Institutes of Medicine, Harvard Medical School, Boston, MA USA
| |
Collapse
|
8
|
Sekizawa K, Nakagawa K, Ichikawa Y, Suwa H, Ozawa M, Momiyama M, Ishibe A, Watanabe J, Ota M, Kato I, Endo I. Relationship between stromal regulatory T cells and the response to neoadjuvant chemotherapy for locally advanced rectal cancer. Surg Today 2021; 52:198-206. [PMID: 34081199 DOI: 10.1007/s00595-021-02311-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/25/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND In addition to the direct power of anticancer drugs, the effectiveness of anticancer therapy depends on the host immune function. The present study investigated whether or not the reduction rate and histological response of preoperative chemotherapy were related to the immune microenvironment surrounding a primary tumor of the rectum. METHODS Sixty-five patients received preoperative chemotherapy followed by resection from 2012 to 2014; all of these patients were retrospectively analyzed. CD3, CD8, and FoxP3 were immunohistochemically examined as markers for T lymphocytes, cytotoxic T lymphocytes, and regulatory T lymphocytes (Treg), respectively. The correlation between the tumor-infiltrating lymphocyte composition and the tumor reduction rate and histological response to neoadjuvant chemotherapy was investigated. RESULTS The average tumor reduction rate was 41.5% ± 18.8%. According to RECIST, 47 patients (72.3%) achieved a partial response (PR), and 1 patient (1.5%) achieved a complete response (CR). Eight patients (12.3%) showed a grade 2 histological response, and 2 (3.1%) showed a grade 3 response. A multivariate analysis demonstrated that a low Treg infiltration in stromal cell areas was significantly associated with the achievement of a PR or CR [odds ratio (OR) 7.69; 95% confidence interval (CI) 1.96-33.33; p < 0.01] and a histological grade 2 or 3 response (OR 11.11; 95% CI 1.37-98.04; p = 0.02). CONCLUSION A low Treg infiltration in the stromal cell areas may be a marker of a good response to neoadjuvant chemotherapy in patients with locally advanced rectal cancer.
Collapse
Affiliation(s)
- Kentaro Sekizawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kazuya Nakagawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Yasushi Ichikawa
- Department of Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hirokazu Suwa
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Mayumi Ozawa
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Masashi Momiyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Atsushi Ishibe
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Jun Watanabe
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Mitsuyoshi Ota
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ikuma Kato
- Department of Molecular Pathology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| |
Collapse
|
9
|
Otegbeye EE, Mitchem JB, Park H, Chaudhuri AA, Kim H, Mutch MG, Ciorba MA. Immunity, immunotherapy, and rectal cancer: A clinical and translational science review. Transl Res 2021; 231:124-138. [PMID: 33307273 PMCID: PMC8016725 DOI: 10.1016/j.trsl.2020.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/28/2020] [Accepted: 12/05/2020] [Indexed: 02/07/2023]
Abstract
Rectal cancer remains a challenging disease to treat. Therapy for locally advanced rectal cancer (LARC), the most frequent presentation, has evolved to include a multimodal approach of radiation, chemotherapy, and surgery. While this approach improves local disease control, the distant recurrence rate is nearly 30% and treatment-related morbidity is substantial, thus underscoring the need for new therapeutic approaches with better efficacy and lower side effects. Immunotherapy could potentially fill this need, but its promise is not yet realized in rectal cancer. In this translational science review, we address what is known about how cytotoxic therapies shape rectal cancer immunity and potentially prime the tumor microenvironment for response to immune checkpoint inhibitors and other immunotherapies. We also address the role of current immunotherapies in colorectal cancer and highlight where novel immunotherapy approaches are currently being evaluated in LARC. Finally, we address important future directions in LARC immunotherapy including the need to define optimal therapeutic sequencing, predictive biomarkers, strategies to limit treatment-related side effects and the potential of gut microbiome manipulation to improve outcomes. In summary, this review provides a framework to guide future research and inform immunotherapy trial design so as to advance rectal cancer care.
Collapse
Affiliation(s)
- Ebunoluwa E Otegbeye
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Jonathan B Mitchem
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri; Surgical Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri
| | - Haeseong Park
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Aadel A Chaudhuri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri; Department of Genetics, Washington University School of Medicine, St. Louis, Missouri; Department of Computer Science & Engineering, Washington University, St. Louis, Missouri
| | - Hyun Kim
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew G Mutch
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri; Department of Surgery, Section of Colorectal Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew A Ciorba
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri; Inflammatory Bowel Diseases Center and the Division of Gastroenterology, Washington University in Saint Louis School of Medicine, St. Louis, Missouri.
| |
Collapse
|
10
|
Hanna CR, O'Cathail SM, Graham J, Adams R, Roxburgh CS. Immune Checkpoint Inhibition as a Strategy in the Neoadjuvant Treatment of Locally Advanced Rectal Cancer. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:86-104. [PMID: 35663532 PMCID: PMC9153256 DOI: 10.36401/jipo-20-31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 06/15/2023]
Abstract
The treatment of locally advanced rectal cancer (LARC) has seen major advances over the past 3 decades, with multimodality treatment now standard of care. Combining surgical resection with radiotherapy and/or chemotherapy can reduce local recurrence from around 20% to approximately 5%. Despite improvements in local control, distant recurrence and subsequent survival rates have not changed. Immune checkpoint inhibitors have improved patient outcomes in several solid tumor types in the neoadjuvant, adjuvant, and advanced disease setting; however, in colorectal cancer, most clinical trials have been performed in the metastatic setting and the benefits confined to microsatellite instability-high tumors. In this article, we review the current preclinical and clinical evidence for using immune checkpoint inhibition in the treatment of LARC and discuss the rationale for specifically exploring the use of this therapy in the neoadjuvant setting. We summarize and discuss relevant clinical trials that are currently in setup and recruiting to test this treatment strategy and reflect on unanswered questions that still need to be addressed within future research efforts.
Collapse
Affiliation(s)
- Catherine R. Hanna
- Cancer Research United Kingdom Clinical Trials Unit, Glasgow, Scotland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Beatson West of Scotland Cancer Centre, Glasgow, Scotland
| | - Séan M. O'Cathail
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Beatson West of Scotland Cancer Centre, Glasgow, Scotland
| | - Janet Graham
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Beatson West of Scotland Cancer Centre, Glasgow, Scotland
| | - Richard Adams
- Centre for Trials Research, Cardiff University and Velindre Cancer Centre, Cardiff, Wales
| | - Campbell S.D. Roxburgh
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland
- Glasgow Royal Infirmary, Glasgow, Scotland
| |
Collapse
|
11
|
Emerging Trends for Radio-Immunotherapy in Rectal Cancer. Cancers (Basel) 2021; 13:cancers13061374. [PMID: 33803620 PMCID: PMC8003099 DOI: 10.3390/cancers13061374] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Rectal cancer is a heterogeneous disease at the genetic and molecular levels, both aspects having major repercussions on the tumor immune contexture. Whilst microsatellite status and tumor mutational load have been associated with response to immunotherapy, presence of tumor-infiltrating lymphocytes is one of the most powerful prognostic and predictive biomarkers. Yet, the majority of rectal cancers are characterized by microsatellite stability, low tumor mutational burden and poor T cell infiltration. Consequently, these tumors do not respond to immunotherapy and treatment largely relies on radiotherapy alone or in combination with chemotherapy followed by radical surgery. Importantly, pre-clinical and clinical studies suggest that radiotherapy can induce a complete reprograming of the tumor microenvironment, potentially sensitizing it for immune checkpoint inhibition. Nonetheless, growing evidence suggest that this synergistic effect strongly depends on radiotherapy dosing, fractionation and timing. Despite ongoing work, information about the radiotherapy regimen required to yield optimal clinical outcome when combined to checkpoint blockade remains largely unavailable. In this review, we describe the molecular and immune heterogeneity of rectal cancer and outline its prognostic value. In addition, we discuss the effect of radiotherapy on the tumor microenvironment, focusing on the mechanisms and benefits of its combination with immune checkpoint inhibitors.
Collapse
|
12
|
Wei Q, Xu Q, Yuan X, Li JJ, Chen L, Luo C, Zhu X, Ying JE. Immunological impact of chemotherapy on the tumor microenvironment in gastric cancer. J Surg Oncol 2021; 123:1708-1715. [PMID: 33684248 DOI: 10.1002/jso.26449] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 02/13/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This study aimed to investigate alterations in pre- and post-neoadjuvant chemotherapy (NACT) tumor-infiltrating immune cells and subsequent evaluation of the predictive and prognostic value of these changes in gastric cancer (GC). METHODS Fifty patients with GC underwent three cycles of S-1 and oxaliplatin (SOX regimen)-NACT. Paired samples from tumor lesions before and after NACT were available for all patients participating in the study. Immunohistochemistry was performed for T cell subsets (CD3+ and CD8+ ) and macrophages (CD68+ and CD163+ ). RESULTS After NACT, the average expression levels of CD3, CD8, CD68, and CD163 were significantly increased (p < .001). However, neither expression levels pre- nor post-chemotherapy correlated with treatment response. Multivariate Cox regression analysis demonstrated that upregulation of CD8/CD3 levels (hazard ratio [HR] = 0.117; 95% confidence interval [CI] = 0.031-0.446; p = 0.002) and CD163 levels after chemotherapy (HR = 2.258; 95% CI = 1.047-4.867; p = 0.038) were independent prognostic factors of overall survival. CONCLUSION Chemotherapy in GC is useful to induce CD3+ and CD8+ T lymphocytes as well as CD68+ and CD163+ macrophages in the tumor microenvironment in combination with its direct cytotoxic effects. These results indicate that chemotherapy may play a role in tumor immune microenvironment remodeling.
Collapse
Affiliation(s)
- Qing Wei
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Qi Xu
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Xing Yuan
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Jing-Jing Li
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Lei Chen
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Cong Luo
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| | - Xiu Zhu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China.,Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China
| | - Jie-Er Ying
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, PR China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, PR China
| |
Collapse
|
13
|
Lumish MA, Cercek A. Immunotherapy for the treatment of colorectal cancer. J Surg Oncol 2021; 123:760-774. [PMID: 33595891 DOI: 10.1002/jso.26357] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibition (ICI) has transformed the management of metastatic colorectal cancer (mCRC) with mismatch-repair deficiency (dMMR) and microsatellite instability (MSI-H), though this constitutes on average less than 5% of mCRC, and ICI is ineffective in preserved MMR/microsatellite stable disease (pMMR/MSS). Here we review the efficacy of ICI in dMMR/MSI-H mCRC, poor response to ICI in pMMR/MSS mCRC, role for ICI in locally advanced disease, biomarkers of response, novel immunotherapies, and future directions in targeting resistance mechanisms.
Collapse
Affiliation(s)
| | - Andrea Cercek
- Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
14
|
Guo J, Yu Z, Sun D, Zou Y, Liu Y, Huang L. Two nanoformulations induce reactive oxygen species and immunogenetic cell death for synergistic chemo-immunotherapy eradicating colorectal cancer and hepatocellular carcinoma. Mol Cancer 2021; 20:10. [PMID: 33407548 PMCID: PMC7786897 DOI: 10.1186/s12943-020-01297-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND FOLFOX is a combinational regimen of folinic acid (FnA, FOL), fluorouracil (5-Fu, F) and oxaliplatin (OxP, OX), and has been long considered as the standard treatment of colorectal cancer (CRC) and hepatocellular carcinoma (HCC). Recent developments of nano delivery systems have provided profound promise for improving anticancer efficacy and alleviating side effects of FOLFOX. Previously, a nanoformulation (termed Nano-Folox) containing OxP derivative and FnA was developed in our laboratory using nanoprecipitation technique. Nano-Folox induced OxP-mediated immunogenic cell death (ICD)-associated antitumor immunity, which significantly suppressed tumor growth in the orthotopic CRC mouse model when administrated in combination with free 5-Fu. METHODS A nanoformulation (termed Nano-FdUMP) containing FdUMP (5-Fu active metabolite) was newly developed using nanoprecipitation technique and used in combination with Nano-Folox for CRC and HCC therapies. RESULTS Synergistic efficacy was achieved in orthotopic CRC and HCC mouse models. It resulted mainly from the fact that Nano-FdUMP mediated the formation of reactive oxygen species (ROS), which promoted the efficacy of ICD elicited by Nano-Folox. In addition, combination of Nano-Folox/Nano-FdUMP and anti-PD-L1 antibody significantly inhibited CRC liver metastasis, leading to long-term survival in mice. CONCLUSION This study provides proof of concept that combination of two nano delivery systems can result in successful FOLFOX-associated CRC and HCC therapies. Further optimization in terms of dosing and timing will enhance clinical potential of this combination strategy for patients.
Collapse
Affiliation(s)
- Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Zhuo Yu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Hepatopathy, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dandan Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yifang Zou
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yun Liu
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
15
|
Priming the tumor immune microenvironment with chemo(radio)therapy: A systematic review across tumor types. Biochim Biophys Acta Rev Cancer 2020; 1874:188386. [PMID: 32540465 DOI: 10.1016/j.bbcan.2020.188386] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Chemotherapy (CT), radiotherapy (RT), and chemoradiotherapy (CRT) are able to alter the composition of the tumor immune microenvironment (TIME). Understanding the effect of these modalities on the TIME could aid in the development of improved treatment strategies. Our aim was to systematically review studies investigating the influence of CT, RT or CRT on different TIME markers. METHODS The EMBASE (Ovid) and PubMed databases were searched until January 2019 for prospective or retrospective studies investigating the dynamics of the local TIME in cancer patients (pts) treated with CT, RT or CRT, with or without targeted agents. Studies could either compare baseline and follow-up specimens - before and after treatment - or a treated versus an untreated cohort. Studies were included if they used immunohistochemistry and/or flow cytometry to assess the TIME. RESULTS In total we included 110 studies (n = 8850 pts), of which n = 89 (n = 6295 pts) compared pre-treatment to post-treatment specimens and n = 25 (n = 2555 pts) a treated versus an untreated cohort (4 studies conducted both comparisons). For several tumor types (among others; breast, cervical, esophageal, ovarian, rectal, lung mesothelioma and pancreatic cancer) remodeling of the TIME was observed, leading to a potentially more immunologically active microenvironment, including one or more of the following: an increase in CD3 or CD8 lymphocytes, a decrease in FOXP3 Tregs and increased PD-L1 expression. Both CT and CRT were able to immunologically alter the TIME. CONCLUSION The TIME of several tumor types is significantly altered after conventional therapy creating opportunities for concurrent or sequential immunotherapy.
Collapse
|
16
|
Drug-Drug Interactions of Irinotecan, 5-Fluorouracil, Folinic Acid and Oxaliplatin and Its Activity in Colorectal Carcinoma Treatment. Molecules 2020; 25:molecules25112614. [PMID: 32512790 PMCID: PMC7321123 DOI: 10.3390/molecules25112614] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
The combination of folinic acid, 5-fluorouracil, oxaliplatin and/or irinotecan (FOLFOXIRI) is the standard of care for metastatic colorectal cancer (CRC). This strategy inhibits tumor growth but provokes drug resistance and serious side effects. We aimed to improve FOLFOXIRI by optimization of the dosing and the sequence of drug administration. We employed an orthogonal array composite design and linear regression analysis to obtain cell line-specific drug combinations for four CRC cell lines (DLD1, SW620, HCT116, LS174T). Our results confirmed the synergy between folinic acid and 5-fluorouracil and additivity, or even antagonism, between the other drugs of the combination. The drug combination administered at clinical doses resulted in significantly higher antagonistic interactions compared to the low-dose optimized drug combination (ODC). We found that the concomitant administration of the optimized drug combination (ODC) was comparatively active to sequential administration. However, the administration of oxaliplatin or the active metabolite of irinotecan seemed to sensitize the cells to the combination of folinic acid and 5-fluorouracil. ODCs were similarly active in non-cancerous cells as compared to the clinically used doses, indicating a lack of reduction of side effects. Interestingly, ODCs were inactive in CRC cells chronically pretreated with FOLFOXIRI, suggesting the occurrence of resistance. We were unable to improve FOLFOXIRI in terms of efficacy or specificity. Improvement of CRC treatment should come from the optimization of targeted drugs and immunotherapy strategies.
Collapse
|
17
|
Park JH, van Wyk H, McMillan DC, Edwards J, Orange C, Horgan PG, Roxburgh CS. Preoperative, biopsy-based assessment of the tumour microenvironment in patients with primary operable colorectal cancer. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2019; 6:30-39. [PMID: 31486287 PMCID: PMC6966701 DOI: 10.1002/cjp2.143] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/18/2019] [Accepted: 08/29/2019] [Indexed: 12/22/2022]
Abstract
The tumour microenvironment (TME) is recognised as an important prognostic characteristic and therapeutic target in patients with colorectal cancer (CRC). However, assessment generally utilises surgically resected specimens, precluding neoadjuvant targeting. The present study investigated the feasibility of intra‐epithelial CD3+ T‐lymphocyte density and tumour stroma percentage (TSP) assessment using preoperative colonoscopic biopsies from 115 patients who had undergone resection of stages I–III CRC, examining the relationship between biopsy and surgically resected specimen‐based assessment, and the relationship with cancer‐specific survival (CSS). High biopsy CD3+ density was associated with high CD3+ density in the invasive margin, cancer stroma and intra‐epithelial compartments of surgically resected specimens (area under the curve > 0.62, p < 0.05 for all) and with high Immunoscore. High biopsy TSP predicted high TSP in resected specimens (p = 0.001). Intra‐class correlation coefficient for both measures was >0.7 (p < 0.001), indicating excellent concordance between individuals. Biopsy CD3+ density (hazard ratio [HR] 0.23, p = 0.002) and TSP (HR 2.23, p = 0.029) were independently associated with CSS; this was comparable to the prognostic value of full section assessment (HR 0.21, p = 0.004, and HR 2.25, p = 0.033 respectively). These results suggest that assessment of the TME is comparable in biopsy and surgically resected specimens from patients with CRC, and biopsy‐based assessment could allow for stratification prior to surgery or commencement of therapy targeting the TME.
Collapse
Affiliation(s)
- James H Park
- Academic Unit of Surgery, School of Medicine Dentistry and Nursing, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Hester van Wyk
- Academic Unit of Surgery, School of Medicine Dentistry and Nursing, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Donald C McMillan
- Academic Unit of Surgery, School of Medicine Dentistry and Nursing, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Joanne Edwards
- Department of Experimental Therapeutics, Institute of Cancer Sciences, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Clare Orange
- NHS Greater Glasgow & Clyde Biorepository, Queen Elizabeth University Hospital, Glasgow, UK
| | - Paul G Horgan
- Academic Unit of Surgery, School of Medicine Dentistry and Nursing, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Campbell Sd Roxburgh
- Academic Unit of Surgery, School of Medicine Dentistry and Nursing, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK.,Department of Experimental Therapeutics, Institute of Cancer Sciences, College of Medicine Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|