Retrospective Study Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Surg. Feb 27, 2023; 15(2): 222-233
Published online Feb 27, 2023. doi: 10.4240/wjgs.v15.i2.222
Efficacy and safety of preoperative immunotherapy in patients with mismatch repair-deficient or microsatellite instability-high gastrointestinal malignancies
Ying-Jie Li, Xin-Zhi Liu, Nan Chen, Ai-Wen Wu, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
Yun-Feng Yao, Gastro-intestinal Ward III, Beijing Cancer Hospital, Beijing 100142, China
Zhong-Wu Li, Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing 100142, China
Xiao-Yan Zhang, Department of Radiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
Xin-Feng Lin, Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
ORCID number: Ying-Jie Li (0000-0001-6091-8911); Yun-Feng Yao (0000-0003-3660-3412); Nan Chen (0000-0002-0085-7472); Zhong-Wu Li (0000-0003-3440-9077); Ai-Wen Wu (0000-0003-1877-7005).
Author contributions: Wu AW contributed to conception and design of the study, draft and final approval of the manuscript; Li YJ contributed to collection of the data, draft the manuscript, study design and statistical analysis; Wu AW, Li YJ, Liu XZ and Yao YF, Li ZW, Zhang XY, Lin XF contributed to quality control of the study especially the surgery part, acquisition of data; All authors approved the final manuscript.
Supported by the National Natural Science Foundation of China, No. 82173156; Beijing Hospitals Authority Clinical Medicine Development of Special Funding, No. ZYLX202116.
Institutional review board statement: This study was approved by the ethics committee of Peking University cancer hospital (approval no. 2022YJZ39).
Informed consent statement: All study participants, or their legal guardian, provided informed written consent prior to study enrollment.
Conflict-of-interest statement: All the authors declare that they have no conflict of interest.
Data sharing statement: Technical appendix, statistical code, and dataset available from the corresponding author at drwuaw@sina.com.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Ai-Wen Wu, MD, PhD, Academic Editor, Director, Doctor, Professor, Surgeon, Teacher, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, China. drwuaw@sina.com
Received: November 29, 2022
Peer-review started: November 29, 2022
First decision: December 27, 2022
Revised: January 8, 2023
Accepted: February 1, 2023
Article in press: February 1, 2023
Published online: February 27, 2023
Processing time: 90 Days and 3.1 Hours

Abstract
BACKGROUND

Programmed death protein (PD)-1 blockade immunotherapy significantly prolongs survival in patients with metastatic mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) gastrointestinal malignancies such gastric and colorectal cancer. However, the data on preoperative immunotherapy are limited.

AIM

To evaluate the short-term efficacy and toxicity of preoperative PD-1 blockade immunotherapy.

METHODS

In this retrospective study, we enrolled 36 patients with dMMR/MSI-H gastrointestinal malignancies. All the patients received PD-1 blockade with or without chemotherapy of CapOx regime preoperatively. PD1 blockade 200 mg was given intravenously over 30 min on day 1 of each 21-d cycle.

RESULTS

Three patients with locally advanced gastric cancer achieved pathological complete response (pCR). Three patients with locally advanced duodenal carcinoma achieved clinical complete response (cCR), followed by watch and wait. Eight of 16 patients with locally advanced colon cancer achieved pCR. All four patients with liver metastasis from colon cancer reached CR, including three with pCR and one with cCR. pCR was achieved in two of five patients with non-liver metastatic colorectal cancer. CR was achieved in four of five patients with low rectal cancer, including three with cCR and one with pCR. cCR was achieved in seven of 36 cases, among which, six were selected for watch and wait strategy. No cCR was observed in gastric or colon cancer.

CONCLUSION

Preoperative PD-1 blockade immunotherapy in dMMR/MSI-H gastrointestinal malignancies can achieve a high CR, especially in patients with duodenal or low rectal cancer, and can achieve high organ function protection.

Key Words: Preoperative; PD-1 blockade; dMMR/MSI-H; Gastrointestinal malignancies

Core Tip: To the best of our knowledge, this retrospective study is one of the few to summarize mismatch repair-deficient/microsatellite instability-high gastric, duodenal, and colorectal cancers for preoperative immunotherapy. The cohort was a sequential case analysis that only one patient was excluded from the cohort because symptoms disappeared after programmed death protein 1 therapy and she refused to examination and further treatment.



INTRODUCTION

Many solid tumors have a mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) ratio, and dMMR/MSI-H subtype, as a unique type of tumor, accounts for 5%–15% of solid tumor patients[1]. However, the proportion of dMMR/MSI-H varies significantly among different tumor types, with a high incidence in colorectal, gastric and endometrial cancer, as well as breast cancer, liver cancer, cholangiocarcinoma, pancreatic cancer and other solid tumors[2]. As a distinguished biomarker, dMMR/MSI-H status can accurately predict the efficacy of immunotherapy for many solid tumors[3,4].

MSI-H subtype is one of the molecular subtypes of gastric or colorectal cancer[5,6]. MSI-H is an independent prognostic factor in gastrointestinal tumors, with good prognosis and resistance to conventional chemoradiotherapy[7,8]. Locally advanced gastrointestinal malignancies or surgically resectable metastatic colorectal cancer require preoperative treatment to improve R0 resection rate. However, many studies[9,10] on locally advanced gastric cancer suggest that the pathological complete response (pCR) rate of conventional preoperative neoadjuvant chemotherapy is not high, and dMMR/MSI-H gastric cancer has a worse response to conventional chemotherapy. As with gastric cancer, pancreaticoduodenectomy (Whipple’s procedure) for duodenal cancer is a major, invasive procedure that needs to be performed even after preoperative chemotherapy. MSI-H colorectal cancer is resistant to conventional chemotherapy[11] and radiotherapy[12], and low advanced rectal cancer requires R0 resection and organ preservation.

In 2015[13] and 2017, Le et al[14] revealed that pembrolizumab [programmed death protein (PD) 1 blockade] achieved a high objective response rate for metastatic dMMR/MSI-H solid malignant tumors, and some patients achieved complete response (CR). In 2017, for the first time, the United States FDA approved pembrolizumab (for immunotherapy of all unresectable, metastatic dMMR/MSI-H solid tumors based on biomarkers instead of tumor types. A series of studies[15-18] on advanced or metastatic dMMR/MSI-h gastrointestinal malignancies such as gastric and colorectal cancer showed that PD1 blockade achieved better therapeutic effects.

This study aimed to evaluate the efficacy and safety of preoperative PD-1 blockade immunotherapy in patients with dMMR/MSI-H locally advanced or metastatic gastrointestinal malignancies.

MATERIALS AND METHODS
Patients

We retrospectively analyzed clinical data of all patients with dMMR/MSI-H gastrointestinal malignancies who received preoperative PD1 blockade immunotherapy with or without chemotherapy in Ward 3, Gastrointestinal Cancer Center, Peking University Cancer Hospital from January 1, 2020 to September 30, 2022. Preoperative PD-1 blockade immunotherapy is recommended for patients with dMMR/MSI-H detected by molecular detection before treatment. This cohort was a series of sequential cases.

The inclusion criteria were: (1) Age ≥ 18 and ≤ 90 years; (2) Eastern Collaborative Oncology Group performance status score 0–2; (3) gastrointestinal malignancies: gastric cancer, duodenal carcinoma, or colorectal cancer; (4) adenocarcinoma or mucous adenocarcinoma confirmed by endoscopic biopsy; (5) clinical stage II–IV according to imaging examinations [spiral computed tomography, magnetic resonance imaging, positron emission tomography–computed tomography (PET-CT) or ultrasound colonoscopy]; and (6) MSI and MMR status confirmed by immumohistochemical staining, polymerase chain reaction (PCR) and next-generation sequencing (NGS). The exclusion criteria included: (1) Initially unresectable metastasis lesion; and (2) diseases of the immune system. This study was approved by the Ethics Committee of Peking University Cancer Hospital (2022YJZ39) and it conformed to the provisions of the Declaration of Helsinki.

Treatment and evaluation

All patients were discussed in a multidisciplinary team conference. All patients received PD1 blockade (PD1 blockade 200 mg intravenously over 30 min on day 1 of each 21-d cycle) preoperative immunotherapy with or without CapOx chemotherapy (oxaliplatin 130 mg/m2 on day 1 and capecitabine 1000 mg/m2 twice daily on d 1–14, repeated every 3 wk).

The primary tumor response was assessed according to the iRECIST criteria[19]. Acute toxicity was graded according to the NCI Common Terminology Criteria for Adverse Events 4.0[20]. After every one or two cycles of neoadjuvant immunotherapy, all patients had complete assessment including computed tomography (CT), magnetic resonance imaging, PET-CT, blood counts, renal biochemistry, hepatobiliary function, thyroid function, cardiac function, and tumor markers (carcinoembryonic antigen and carbohydrate antigen 19-9) to evaluate the general condition and treatment response. The determination of clinical complete response (cCR) was based on Memorial Sloan Kettering Cancer Center standard[21] and International Watch & Wait Database[22]. The cCR was defined as no evidence of residual tumor determined by rectum MRI, abdomen/pelvis CT and chest CT, endoscopic physical examination, nomarl CEA and/or digital rectal exam. Pathological staging was based on the 8th edition of the American Joint Committee on Cancer TNM staging system[23]. Post-treatment response was assessed by NCCN grading: 0 = complete response (ypCR) with no detectable cancer cells; 1 = major response with few residual cancer cells; 2 = partial response; 3 = no or very little response[24].

Postoperative complications were classified according to the Clavien–Dindo classification[24].

Follow-up and statistical analysis

After initial treatment, all patients were re-examined every 3 mo for the first 2 years, 6-mo intervals for the next 3 years, and yearly thereafter. Clinical data were obtained by laboratory examination records, imaging examination records, pathological examination records, and medical records review. Survival data were obtained from outpatient clinical visit or telephone interview. Analyses were conducted using SPSS for Windows version 19.0. Frequencies and percentages were calculated for categorical variables, and means and standard deviations for continuous variables.

RESULTS
Patient characteristics

From January 1, 2020 to September 30, 2022, 37 patients with dMMR/MSI-H gastrointestinal malignancies finished preoperative PD1 blockade immunotherapy, and one patient with locally advanced colon cancer refused re-examination and treatment after symptom resolution. By September 30, 2022, 36 patients completed PD1 blockade immunotherapy and the treatment results were analyzed.

The clinical features are shown in Tables 1 and 2. Twenty-six men (26/36, 72.2%) and 10 women (10/36, 27.8%) were enrolled and the median age was 52 (26–87) years. Lynch syndrome was diagnosed in 16 patients. Three patients were diagnosed with initially unresectable locally advanced gastric cancer, three with locally advanced duodenal carcinoma, and 30 with colorectal cancer. Among the 30 cases of colorectal cancer, 21 were locally advanced and nine were surgically resectable metastatic colorectal cancer. Twenty-seven patients received single PD-1 blockade as preoperative therapy, and nine with PD1 blockade and CapOx regimen. Of these patients, nine were initially treated with CapOx and two with radiotherapy, which was ineffective and switched to immunotherapy.

Table 1 Patient, tumor and treatment characteristics.
All cases
N (%)
Sex, n (%)
Male26 (72.2)
Female10 (27.8)
Age, yr (median)52.0
ECOG
0 2 (5.6)
1 30 (83.3)
2 4 (11.1)
Tumor location, n (%)
Stomach3 (8.3)
Duodenum3 (8.3)
Colorectal cancer30 (83.3)
Clinical TNM stage, n (%)
II-III27(75.0)
IV9 (25.0)
Lynch syndrome
Yes16 (44.4)
No20 (55.6)
No clear0 (0.0)
MMR and MSI status
Consistent30 (83.3)
Inconsistent6 (16.7)
Table 2 Details of the 36 patients with neoadjuvant programmed death protein 1 blockade immunotherapy.
No.Tumor locationClinical stageMMR statusMSI statusCourses of ICBCombined treatmentSurgeryPathological stage
1StomachT4aN2M0dMMRMSI-HSintilimab × 6CapeOXTotal gastrectomyPCR
2StomachT4aN2M0dMMRMSI-HSintilimab × 5CapeOXTotal gastrectomyPCR
3StomachT4aN2M0dMMRMSI-HSintilimab × 3CapeOXTotal gastrectomyPCR
4DuodenumT3N0M0dMMRMSI-HPembrolizumab × 4-cCR-
5DuodenumT3N0M0dMMRMSI-HPembrolizumab × 6-cCR-
6DuodenumT4aN0M0dMMRMSI-LSintilimab × 8CapeOXcCR-
7LACCT4aN+M0dMMRMSI-HPembrolizumab × 2-ColectomyPCR
8LACCT3N+M0dMMRMSI-HSintilimab × 2-ColectomyypT3N0
9LACCT4bN2M0dMMRMSI-HPembrolizumab × 2-ColectomyPCR
10LACCT3N+M0dMMRMSI-HCamrelizumab × 4CapeOXColectomyPCR
11LACCT4bN+M0dMMRMSI-HPembrolizumab × 1-ColectomyypT3N0
12LACCT3N+M0dMMRMSI-HCamrelizumab × 3-ColectomyypT2N0
13LACCT4bN2bM0dMMRMSI-HSintilimab × 4-ColectomyypT1N1b
14LACCT4bN+M0dMMRMSI-HPembrolizumab × 2-ColectomyPCR
15LACCT4aN+M0dMMRMSSSintilimab × 3CapeOXColectomyypT3N0
16LACCT4aN+M0dMMRMSI-HToripalimab × 8-ColectomyypT2N0
17LACCT3N0M0dMMRMSI-HPembrolizumab × 4-ColectomyypT3N0
18LACCT4bN2aM0dMMRMSI-HSintilimab × 5-ColectomyPCR
19LACCT4bN2bM0dMMRMSI-HSintilimab × 3-ColectomyPCR
20LACCT4aN2bM0dMMRMSSPembrolizumab × 3-ColectomyypT3N1a
21LACCT3N2aM0pMMRMSI-HPembrolizumab × 3-ColectomyPCR
22LACCcT4aN2M0dMMRMSI-HPembrolizumab × 4-ColectomyPCR
23Low rectumT3N1M0dMMRMSI-HPembrolizumab × 1-CCR-
24Low rectumT3N2M0dMMRMSI-HNivolumab × 3CapeOXLARPCR
25Low rectumT3N+M0pMMRMSI-HSintilimab × 4-LARypT3N0
26Low rectumT3N+M0dMMRMSI-HPembrolizumab × 2-CCR-
27Low rectumT3N2M0pMMRMSI-HCamrelizumab × 3CapeOXCCRPCR
28CLMM1adMMRMSI-HSintilimab × 6CapeOXHepatectomyPCR
29CLMT4bN2M1adMMRMSI-HSintilimab × 4CapeOXColectomyHepatectomyypT4bN1aM0
30CLMM1adMMRMSI-HSintilimab × 26-CCR -
31CLMT4aN2M1adMMRMSI-HPembrolizumab × 4-ColectomyHepatectomyPCR
32RectumT4bN2M1cdMMRMSI-HPembrolizumab × 1-ColectomyypT4bN2aM1c
33RectumT4bN2M1adMMRMSI-HSintilimab × 6-HartmannPCR
34ColonT4bN2M1cdMMRMSI-HTislelizumab × 8-ColectomyypT0N0M1c
35ColonT4bN2M1cdMMRMSI-HSintilimab × 5CapeOXColectomyypT3N0M1c
36ColonT4aN1M1cdMMRMSI-HSintilimab × 5-ColectomyPCR
Efficacy

With a median interval of 2 wk after neoadjuvant immunotherapy (range: 1–4 wk), all patients received imaging evaluation (Table 2). cCR was achieved in seven patients (patient 4, 5, 6, 23, 26, 27, 30), among which six (patient 4, 5, 6, 23, 26, 30) were selected for wait and watch strategy. No cCR was observed in gastric or colon cancer. Thirty patients underwent surgery, including nine laparoscopic operations and 21 Laparotomies. Twenty-nine patients achieved R0 resection according to the postoperative pathological diagnosis (Table 2).

Three patients with locally advanced gastric cancer achieved pathological CR (pCR). Three patients with locally advanced duodenal carcinoma achieved cCR and then watch and wait (Video 1). Eight of 16 patients with locally advanced colon cancer achieved pCR. All four patients with liver metastasis from colon cancer achieved CR, including three pCR and one cCR. pCR was achieved in two of five patients with non-liver metastases from colorectal cancer. CR was achieved in four of five patients with low rectal cancer, including three with cCR and one with pCR. The CR (cCR and pCR) rate was 58.3% (21/36).

No adjuvant therapy was performed after pCR was achieved. Six patients received 2–10 cycles of adjuvant mono-immunotherapy after reaching cCR. The median follow-up was 9.4 (1–29) mo, and final follow-up time was September 30, 2022. pCR and cCR patients had no recurrence during follow-up. No patient died during follow-up.

Safety

Preoperative immunotherapy was well tolerated and there was no interruption caused by immunotherapy-related toxicity. Two patients had grade 3 toxicity that presented as hypothyroidism and hematochezia, and eight patients had grade 1 or 2 toxicity including hypothyroidism, thyroid toxicity, rash, fatigue, reactive cutaneous capillary endothelial proliferation (Table 3). No surgery was delayed, but four patients experienced emergency surgery, of which two had perforated tumor, and two had colonic obstruction. There was no 30-d mortality. Four patients had grade 1 or 2 postoperative complications, including two grade 2 chylous fistula, two grade 2 anastomotic leak, and one grade 1 fever. One patient (case 24) underwent emergency surgery for intestinal obstruction, and received a second operation for postoperative abdominal bleeding (grade 3). All patients recovered well after conservative treatment and only one (anastomotic leak) stayed in hospital > 2 wk after surgery.

Table 3 Adverse events.
Adverse events
No. of patients (%)
No. of patients (%)
No. of patients (%)
Immuno-related Grade 1Grade 2Grade 3
Any4 (11.1)4 (11.1)2 (5.6)
Dermatologic
Rash 2 (5.6)0 (0)0 (0)
RCCEP1 (2.80)0 (0)0 (0)
Endocrine
Hypothyroidism2 (5.6)2 (5.6)1 (2.80)
Hyperglycemia0 (0)0 (0)0 (0)
Fatigue3 (8.3)4(11.1)0 (0)
Surgery-related
Any1 (2.80)4(11.1)1 (2.80)
Chylous fistula0 (0)2 (5.6)0 (0)
Anastomosis leakage0 (0)2 (5.6)0 (0)
Fever1 (2.80)0 (0)0 (0)
Intra-abdominal hemorrhage0 (0)0 (0)1 (2.80)
DISCUSSION

This retrospective study is one of the few to summarize dMMR/MSI-H gastric, duodenal and colorectal cancers for preoperative immunotherapy. The cohort was a sequential case analysis and only one patient was excluded because her symptoms disappeared after PD1 therapy and she refused further examination and treatment.

The GERCOR NEONIPIGA study[25] reported neoadjuvant nivolumab plus ipilimumab in patients with localized dMMR/MSI-H esophagogastric adenocarcinoma showed a pCR rate of 58.6%. The NICHE study[26] and PICC study[27] of dMMR/MSI-H colon cancer showed a pCR rate of 60% and 65%, respectively. Cercek et al[28] reported 14 patients with dMMR/MSI-H rectal cancer using dostarlimab (PD1 blockade); all of whom achieved cCR and avoided surgical treatment.

MSI-H/dMMR tumors are currently the best predictive markers for cancer immunotherapy, but MSI-H tumors are also heterogeneous, and show different incidence, differences in clinical and radiographic responses to immunotherapy in different solid tumors, primary immunotherapy resistance in some patients, and heterogeneity of different stages of the same tumor[1,29-31]. In our cohort three patients with locally advanced gastric cancer achieved pCR. Three patients with locally advanced duodenal carcinoma evaluated as cCR received the watch and wait strategy. Eight of 16 patients with locally advanced colon cancer achieved pCR. All four patients with liver metastatic lesions from colon cancer reached CR. pCR was achieved in two of five patients with non-liver metastatic colorectal cancer. CR was achieved in four of five patients with low rectal cancer, including three with cCR and one with pCR. No cCR was observed in gastric or colon cancer. Duodenal and low rectal cancers have a high cCR rate, and do not require surgery and achieve organ preservation. Based on such high pCR and cCR rates, some patients can avoid surgery. Eleven of 36 patients in our study underwent routine preoperative chemotherapy and/or radiotherapy, which was unnecessary. Neoadjuvant therapy should be effective, moderate and accurate based on the treatment target. It is necessary to determine the molecular status of patients before treatment.

Different from the prospective clinical study, patients in our cohort underwent surgery and achieved R0 resection on imaging examination. Five patients achieved cCR or pCR within one or two cycles, and 15 patients who achieved pCR did not receive postoperative adjuvant therapy. None of the patients with pCR or cCR had recurrence or metastasis during follow-up. The Keynote 016[13], Keynote 177[2] and CHECKMATE 142[16] studies showed that if MSI-H tumors were highly sensitive to PD1 inhibitors or CTLA4 (cytotoxic T lymphocyte-associated antigen-4, CTLA-4) antibody, the response was rapid, and there was a significant tail effect of immunotherapy after long follow-up. Because of the inaccuracy of conventional imaging in the assessment of immunotherapy, the development of new tests that reflect the pathological response (e.g., metabolic imaging) or molecular features (e.g., liquid biopsy) is needed to better assess the response to treatment[32]. For MSI-H tumors sensitive to PD1 immunotherapy, early and short-term PD1 treatment can achieve a curative effect. It is necessary to clarify the characteristics and immune microenvironment of MSI-H tumors.

Our study and others have shown that preoperative immunotherapy is safe for most patients[33,34]. However, we have seen that there are some hidden dangers in the use of immunotherapy for such patients before surgery, such as stenosis, obstruction and perforation during the treatment of colon cancer. Previous studies[35,36] of neoadjuvant chemotherapy have suggested that obstruction and perforation reflect the poor effect of neoadjuvant therapy and are associated with tumor progression and poor prognosis. Due to immune cell infiltration and other reasons, many patients did not observe tumor remission on imaging-maintained stability or even some enlargement, but pathological examination will find a large number of necrosis tumors and inflammatory immune response[37]. Our study also observed this phenomenon. Our cohort reported four patients with MSI-H colon cancer with obstruction and perforation during PD1 treatment; all of whom had some tumor pathological response after emergency surgery, including one who achieved pCR and three with pathological ratings of TRG2–3. We also observed five patients with regional lymph node enlargement during treatment. Although no trace of metastasis was found after lymph node dissection, it increased the difficulty of surgery and was prone to complications of lymphatic leakage. The incidence of thyroid dysfunction was 22% and three patients required long-term oral thyroxine replacement therapy.

Many studies[2,3,14-18] have reported that 10%–40% of patients with dMMR/MSI-H colorectal cancer developed primary drug resistance when receiving immunotherapy. Cohenet al[38] suggested that primary resistance to PD-1 blockades in dMMR/MSI-H metastatic colorectal cancer can largely be attributed to misdiagnosis of MMR/MSI status. In our cohort, 16.7% (6/36) of patients had MMR and MSI discordance[39,40]. We found that this discordance was test related and in some patients it was related to examination methods, tissue volume, tumor heterogeneity, and examination quality control. Six patients were rediagnosed with MMR and MSI status, and three were found to be consistent (Table 4). Our study and many others have found that intrinsic resistance exists even when MSI is consistent with MMR status[39,40]. Wang et al[41] found that combination of AKT1 and CDH1 mutations predicted primary resistance to immunotherapy in dMMR/MSI-H gastrointestinal cancer.

Table 4 Details of the 6 patients with inconsistency of dMMR and MSI-H.
No.Tumor locationClinical stageHeredityMMR (IHC)biopsyMMR (NGS) biopsyMSI (PCR) biopsyMSI (NGS) biopsyMMR (IHC) surgeryMMR (NGS) surgeryMMR (PCR) surgeryMMR (NGS) surgeryReason
6DuodenumT4aN+M0LynchdMMRdMMRMSI-LMSI-L-
15ColonT4aN+M0NodMMRpMMRMSSMSSpMMRpMMRMSSMSSTumor heterogeneity
20ColonT4aN2bM0NodMMRdMMRMSSMSSdMMRdMMRMSI-HMSI-HLow tumor content
21.ColonT3N2aM0NopMMRdMMRMSI-HMSI-HOnly MSH3 mt
25RectumT3N+M0NopMMRpMMRMSSMSI-HpMMRpMMRMSSMSI-HDDR mt
27RectumT3N2M0NopMMRpMMRMSSMSI-HDDR mt

The limitations of this study included its small sample size, retrospective design, short follow-up time, and different neoadjuvant regimens and cycles. However, our study was a real-world clinical study in patients who required preoperative immunotherapy, and the group was a continuous case cohort, with the exception of one patient who declined to be enrolled because of resolution of symptoms. This study compared preoperative immunotherapy for dMMR/MSI-H in different gastrointestinal tumors, and showed that specific treatment strategies could be used for different tumor sites. It was encouraging to find that a high proportion of dMMR/MSI-H duodenal and low rectal cancers did not require surgery after preoperative immunotherapy, and this treatment strategy deserves further investigation.

CONCLUSION

Our study demonstrated that preoperative PD-1 blockade immunotherapy with or without chemotherapy achieved significant results and acceptable adverse events in dMMR/MSI-H gastrointestinal malignancies. Some low cases of low rectal cancer or duodenal cancer can achieve cCR and avoid surgery and achieve organ preservation. Larger clinical trials are needed to conform our results.

ARTICLE HIGHLIGHTS
Research background

Neoadjuvant programmed death protein (PD)-1 blockade immunotheapy has been sufficiently applied in a variety of cancers, but was rare in metastatic mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) gastrointestinal malignancies. Since the NICHE Study have showed their inspiring results that neoadjuvant immunotherapy was an efficient and safe method to improve colon cancer patients’ outcome, NCCN guideline then recommend immune checkpoint inhibitors to cT4b gastric or colorectal cancer. However, whether preoperative immunotherapy can expand to other stage gastrointestinal malignancies is still unknown.

Research motivation

We performed this study among 36 initially surgical resected difficult dMMR/MSI-H gastrointestinal malignancies such gastric cancer, duodenal cancer and colorectal cancer patients who received preoperative PD-1 blockade immunotherapy followed by surgery in order to investigate if the indication of preoperative immunotherapy can expand to initially surgical resected difficult dMMR/MSI-H gastrointestinal malignancies and evaluate the safety and efficacy.

Research objectives

To the best of our knowledge, this retrospective study is one of the few to summarize dMMR/MSI-H gastric, duodenal, and colorectal cancers for preoperative immunotherapy. The cohort was a sequential case analysis that only one patient was excluded from the cohort because symptoms disappeared after PD1 therapy and she refused to examination and further treatment.

Research methods

The limitations of this study included its small sample size, retrospective design, short follow-up time, and different neoadjuvant regimens and cycles. However, our study was a real-world clinical study in patients who required preoperative immunotherapy, and the group was a continuous case cohort, with the exception of one patient who declined to be enrolled because of resolution of symptoms.

Research results

Our study demonstrated that preoperative PD-1 blockade immunotherapy with or without chemotherapy could achieve significant effect and acceptable adverse events in dMMR/MSI-H gastrointestinal malignancies.

Research conclusions

Our study demonstrated that preoperative PD-1 blockade immunotherapy with or without chemotherapy could achieve significant effect and acceptable adverse events in dMMR/MSI-H gastrointestinal malignancies. Some low rectal cancer or duodenal cancer can achieve clinical complete response and avoid surgery to achieve organ preservation. Large sample clinical trials are needed.

Research perspectives

This study compared preoperative immunotherapy for dMMR/MSI-H in different gastrointestinal tumors, and showed that specific treatment strategies could be used for different tumor sites. It was encouraging to find that a high proportion of dMMR/MSI-H duodenal and low rectal cancers did not require surgery after preoperative immunotherapy, and this treatment strategy deserves further investigation.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Immunology

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): D, D

Grade E (Poor): 0

P-Reviewer: Cerwenka H, Austria; Milani L, Italy; Rabago LR, Spain S-Editor: Liu JH L-Editor: A P-Editor: Liu JH

References
1.  Lorenzi M, Amonkar M, Zhang J, Mehta S, Liaw KL. Epidemiology of Microsatellite Instability High (MSI-H) and Deficient Mismatch Repair (dMMR) in Solid Tumors: A Structured Literature Review. J Oncol. 2020;2020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
2.  Hause RJ, Pritchard CC, Shendure J, Salipante SJ. Classification and characterization of microsatellite instability across 18 cancer types. Nat Med. 2016;22:1342-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 481]  [Cited by in F6Publishing: 642]  [Article Influence: 80.3]  [Reference Citation Analysis (0)]
3.  André T, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, de la Fouchardiere C, Rivera F, Elez E, Bendell J, Le DT, Yoshino T, Van Cutsem E, Yang P, Farooqui MZH, Marinello P, Diaz LA Jr; KEYNOTE-177 Investigators. Pembrolizumab in Microsatellite-Instability-High Advanced Colorectal Cancer. N Engl J Med. 2020;383:2207-2218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 962]  [Cited by in F6Publishing: 1450]  [Article Influence: 362.5]  [Reference Citation Analysis (0)]
4.  Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, Geva R, Gottfried M, Penel N, Hansen AR, Piha-Paul SA, Doi T, Gao B, Chung HC, Lopez-Martin J, Bang YJ, Frommer RS, Shah M, Ghori R, Joe AK, Pruitt SK, Diaz LA Jr. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol. 2020;38:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 942]  [Cited by in F6Publishing: 1678]  [Article Influence: 335.6]  [Reference Citation Analysis (0)]
5.  Guinney J, Dienstmann R, Wang X, de Reyniès A, Schlicker A, Soneson C, Marisa L, Roepman P, Nyamundanda G, Angelino P, Bot BM, Morris JS, Simon IM, Gerster S, Fessler E, De Sousa E Melo F, Missiaglia E, Ramay H, Barras D, Homicsko K, Maru D, Manyam GC, Broom B, Boige V, Perez-Villamil B, Laderas T, Salazar R, Gray JW, Hanahan D, Tabernero J, Bernards R, Friend SH, Laurent-Puig P, Medema JP, Sadanandam A, Wessels L, Delorenzi M, Kopetz S, Vermeulen L, Tejpar S. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21:1350-1356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3408]  [Cited by in F6Publishing: 3180]  [Article Influence: 353.3]  [Reference Citation Analysis (0)]
6.  Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4230]  [Cited by in F6Publishing: 4486]  [Article Influence: 448.6]  [Reference Citation Analysis (2)]
7.  Yang G, Zheng RY, Jin ZS. Correlations between microsatellite instability and the biological behaviour of tumours. J Cancer Res Clin Oncol. 2019;145:2891-2899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 78]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
8.  Popat S, Hubner R, Houlston RS. Systematic review of microsatellite instability and colorectal cancer prognosis. J Clin Oncol. 2005;23:609-618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1267]  [Cited by in F6Publishing: 1296]  [Article Influence: 68.2]  [Reference Citation Analysis (1)]
9.  Kim SY, Choi YY, An JY, Shin HB, Jo A, Choi H, Seo SH, Bang HJ, Cheong JH, Hyung WJ, Noh SH. The benefit of microsatellite instability is attenuated by chemotherapy in stage II and stage III gastric cancer: Results from a large cohort with subgroup analyses. Int J Cancer. 2015;137:819-825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 77]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
10.  Pietrantonio F, Miceli R, Raimondi A, Kim YW, Kang WK, Langley RE, Choi YY, Kim KM, Nankivell MG, Morano F, Wotherspoon A, Valeri N, Kook MC, An JY, Grabsch HI, Fucà G, Noh SH, Sohn TS, Kim S, Di Bartolomeo M, Cunningham D, Lee J, Cheong JH, Smyth EC. Individual Patient Data Meta-Analysis of the Value of Microsatellite Instability As a Biomarker in Gastric Cancer. J Clin Oncol. 2019;37:3392-3400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 251]  [Cited by in F6Publishing: 256]  [Article Influence: 51.2]  [Reference Citation Analysis (0)]
11.  Seymour MT, Morton D. FOxTROT: An international randomised controlled trial in 1052 patients (pts) evaluating neoadjuvant chemotherapy (NAC) for colon cancer. J Clin Oncol. 2019;37:3504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 105]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
12.  Hasan S, Renz P, Wegner RE, Finley G, Raj M, Monga D, McCormick J, Kirichenko A. Microsatellite Instability (MSI) as an Independent Predictor of Pathologic Complete Response (PCR) in Locally Advanced Rectal Cancer: A National Cancer Database (NCDB) Analysis. Ann Surg. 2020;271:716-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 87]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
13.  Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Duffy SM, Goldberg RM, de la Chapelle A, Koshiji M, Bhaijee F, Huebner T, Hruban RH, Wood LD, Cuka N, Pardoll DM, Papadopoulos N, Kinzler KW, Zhou S, Cornish TC, Taube JM, Anders RA, Eshleman JR, Vogelstein B, Diaz LA Jr. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509-2520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6096]  [Cited by in F6Publishing: 6780]  [Article Influence: 753.3]  [Reference Citation Analysis (0)]
14.  Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, Lu S, Kemberling H, Wilt C, Luber BS, Wong F, Azad NS, Rucki AA, Laheru D, Donehower R, Zaheer A, Fisher GA, Crocenzi TS, Lee JJ, Greten TF, Duffy AG, Ciombor KK, Eyring AD, Lam BH, Joe A, Kang SP, Holdhoff M, Danilova L, Cope L, Meyer C, Zhou S, Goldberg RM, Armstrong DK, Bever KM, Fader AN, Taube J, Housseau F, Spetzler D, Xiao N, Pardoll DM, Papadopoulos N, Kinzler KW, Eshleman JR, Vogelstein B, Anders RA, Diaz LA Jr. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357:409-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3799]  [Cited by in F6Publishing: 4514]  [Article Influence: 644.9]  [Reference Citation Analysis (0)]
15.  Le DT, Kim TW, Van Cutsem E, Geva R, Jäger D, Hara H, Burge M, O'Neil B, Kavan P, Yoshino T, Guimbaud R, Taniguchi H, Elez E, Al-Batran SE, Boland PM, Crocenzi T, Atreya CE, Cui Y, Dai T, Marinello P, Diaz LA Jr, André T. Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: KEYNOTE-164. J Clin Oncol. 2020;38:11-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 590]  [Article Influence: 118.0]  [Reference Citation Analysis (0)]
16.  Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, Desai J, Hill A, Axelson M, Moss RA, Goldberg MV, Cao ZA, Ledeine JM, Maglinte GA, Kopetz S, André T. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017;18:1182-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1775]  [Cited by in F6Publishing: 1909]  [Article Influence: 272.7]  [Reference Citation Analysis (0)]
17.  Diaz LA Jr, Shiu KK, Kim TW, Jensen BV, Jensen LH, Punt C, Smith D, Garcia-Carbonero R, Benavides M, Gibbs P, de la Fourchardiere C, Rivera F, Elez E, Le DT, Yoshino T, Zhong WY, Fogelman D, Marinello P, Andre T; KEYNOTE-177 Investigators. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol. 2022;23:659-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 317]  [Article Influence: 158.5]  [Reference Citation Analysis (0)]
18.  Janjigian YY, Shitara K, Moehler M, Garrido M, Salman P, Shen L, Wyrwicz L, Yamaguchi K, Skoczylas T, Campos Bragagnoli A, Liu T, Schenker M, Yanez P, Tehfe M, Kowalyszyn R, Karamouzis MV, Bruges R, Zander T, Pazo-Cid R, Hitre E, Feeney K, Cleary JM, Poulart V, Cullen D, Lei M, Xiao H, Kondo K, Li M, Ajani JA. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial. Lancet. 2021;398:27-40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1201]  [Cited by in F6Publishing: 1394]  [Article Influence: 464.7]  [Reference Citation Analysis (0)]
19.  Seymour L, Bogaerts J, Perrone A, Ford R, Schwartz LH, Mandrekar S, Lin NU, Litière S, Dancey J, Chen A, Hodi FS, Therasse P, Hoekstra OS, Shankar LK, Wolchok JD, Ballinger M, Caramella C, de Vries EGE; RECIST working group. iRECIST: guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017;18:e143-e152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1521]  [Cited by in F6Publishing: 1509]  [Article Influence: 215.6]  [Reference Citation Analysis (0)]
20.  Chen AP, Setser A, Anadkat MJ, Cotliar J, Olsen EA, Garden BC, Lacouture ME. Grading dermatologic adverse events of cancer treatments: the Common Terminology Criteria for Adverse Events Version 4.0. J Am Acad Dermatol. 2012;67:1025-1039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 216]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
21.  Smith JJ, Strombom P, Chow OS, Roxburgh CS, Lynn P, Eaton A, Widmar M, Ganesh K, Yaeger R, Cercek A, Weiser MR, Nash GM, Guillem JG, Temple LKF, Chalasani SB, Fuqua JL, Petkovska I, Wu AJ, Reyngold M, Vakiani E, Shia J, Segal NH, Smith JD, Crane C, Gollub MJ, Gonen M, Saltz LB, Garcia-Aguilar J, Paty PB. Assessment of a Watch-and-Wait Strategy for Rectal Cancer in Patients With a Complete Response After Neoadjuvant Therapy. JAMA Oncol. 2019;5:e185896.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 329]  [Article Influence: 65.8]  [Reference Citation Analysis (0)]
22.  van der Valk MJM, Hilling DE, Bastiaannet E, Meershoek-Klein Kranenbarg E, Beets GL, Figueiredo NL, Habr-Gama A, Perez RO, Renehan AG, van de Velde CJH; IWWD Consortium. Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the International Watch & Wait Database (IWWD): an international multicentre registry study. Lancet. 2018;391:2537-2545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 497]  [Cited by in F6Publishing: 619]  [Article Influence: 103.2]  [Reference Citation Analysis (0)]
23.  Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, Grem JL, Grothey A, Hochster HS, Hoffe S, Hunt S, Kamel A, Kirilcuk N, Krishnamurthi S, Messersmith WA, Meyerhardt J, Mulcahy MF, Murphy JD, Nurkin S, Saltz L, Sharma S, Shibata D, Skibber JM, Sofocleous CT, Stoffel EM, Stotsky-Himelfarb E, Willett CG, Wuthrick E, Gregory KM, Gurski L, Freedman-Cass DA. Rectal Cancer, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16:874-901.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 447]  [Cited by in F6Publishing: 620]  [Article Influence: 124.0]  [Reference Citation Analysis (0)]
24.  Clavien PA, Barkun J, de Oliveira ML, Vauthey JN, Dindo D, Schulick RD, de Santibañes E, Pekolj J, Slankamenac K, Bassi C, Graf R, Vonlanthen R, Padbury R, Cameron JL, Makuuchi M. The Clavien-Dindo classification of surgical complications: five-year experience. Ann Surg. 2009;250:187-196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6210]  [Cited by in F6Publishing: 7710]  [Article Influence: 514.0]  [Reference Citation Analysis (0)]
25.  André T, Tougeron D, Piessen G, de la Fouchardière C, Louvet C, Adenis A, Jary M, Tournigand C, Aparicio T, Desrame J, Lièvre A, Garcia-Larnicol ML, Pudlarz T, Cohen R, Memmi S, Vernerey D, Henriques J, Lefevre JH, Svrcek M. Neoadjuvant Nivolumab Plus Ipilimumab and Adjuvant Nivolumab in Localized Deficient Mismatch Repair/Microsatellite Instability-High Gastric or Esophagogastric Junction Adenocarcinoma: The GERCOR NEONIPIGA Phase II Study. J Clin Oncol. 2023;41:255-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 132]  [Article Influence: 132.0]  [Reference Citation Analysis (0)]
26.  Chalabi M, Fanchi LF, Dijkstra KK, Van den Berg JG, Aalbers AG, Sikorska K, Lopez-Yurda M, Grootscholten C, Beets GL, Snaebjornsson P, Maas M, Mertz M, Veninga V, Bounova G, Broeks A, Beets-Tan RG, de Wijkerslooth TR, van Lent AU, Marsman HA, Nuijten E, Kok NF, Kuiper M, Verbeek WH, Kok M, Van Leerdam ME, Schumacher TN, Voest EE, Haanen JB. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers. Nat Med. 2020;26:566-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 341]  [Cited by in F6Publishing: 720]  [Article Influence: 180.0]  [Reference Citation Analysis (0)]
27.  Hu H, Kang L, Zhang J, Wu Z, Wang H, Huang M, Lan P, Wu X, Wang C, Cao W, Hu J, Huang Y, Huang L, Shi L, Cai Y, Shen C, Ling J, Xie X, He X, Dou R, Zhou J, Ma T, Zhang X, Luo S, Deng W, Ling L, Liu H, Deng Y. Neoadjuvant PD-1 blockade with toripalimab, with or without celecoxib, in mismatch repair-deficient or microsatellite instability-high, locally advanced, colorectal cancer (PICC): a single-centre, parallel-group, non-comparative, randomised, phase 2 trial. Lancet Gastroenterol Hepatol. 2022;7:38-48.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 115]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
28.  Cercek A, Lumish M, Sinopoli J, Weiss J, Shia J, Lamendola-Essel M, El Dika IH, Segal N, Shcherba M, Sugarman R, Stadler Z, Yaeger R, Smith JJ, Rousseau B, Argiles G, Patel M, Desai A, Saltz LB, Widmar M, Iyer K, Zhang J, Gianino N, Crane C, Romesser PB, Pappou EP, Paty P, Garcia-Aguilar J, Gonen M, Gollub M, Weiser MR, Schalper KA, Diaz LA Jr. PD-1 Blockade in Mismatch Repair-Deficient, Locally Advanced Rectal Cancer. N Engl J Med. 2022;386:2363-2376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 595]  [Cited by in F6Publishing: 637]  [Article Influence: 318.5]  [Reference Citation Analysis (0)]
29.  Vilar E, Gruber SB. Microsatellite instability in colorectal cancer-the stable evidence. Nat Rev Clin Oncol. 2010;7:153-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 660]  [Cited by in F6Publishing: 646]  [Article Influence: 46.1]  [Reference Citation Analysis (0)]
30.  Lee MKC, Loree JM. Current and emerging biomarkers in metastatic colorectal cancer. Curr Oncol. 2019;26:S7-S15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
31.  Li SKH, Martin A. Mismatch Repair and Colon Cancer: Mechanisms and Therapies Explored. Trends Mol Med. 2016;22:274-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 122]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
32.  Cottrell TR, Thompson ED, Forde PM, Stein JE, Duffield AS, Anagnostou V, Rekhtman N, Anders RA, Cuda JD, Illei PB, Gabrielson E, Askin FB, Niknafs N, Smith KN, Velez MJ, Sauter JL, Isbell JM, Jones DR, Battafarano RJ, Yang SC, Danilova L, Wolchok JD, Topalian SL, Velculescu VE, Pardoll DM, Brahmer JR, Hellmann MD, Chaft JE, Cimino-Mathews A, Taube JM. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC). Ann Oncol. 2018;29:1853-1860.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 320]  [Article Influence: 64.0]  [Reference Citation Analysis (0)]
33.  Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, Horn L, Drake CG, Pardoll DM, Chen L, Sharfman WH, Anders RA, Taube JM, McMiller TL, Xu H, Korman AJ, Jure-Kunkel M, Agrawal S, McDonald D, Kollia GD, Gupta A, Wigginton JM, Sznol M. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443-2454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8900]  [Cited by in F6Publishing: 9508]  [Article Influence: 792.3]  [Reference Citation Analysis (0)]
34.  Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, Shen C, Duma N, Vera Aguilera J, Chintakuntlawar A, Price KA, Molina JR, Pagliaro LC, Halfdanarson TR, Grothey A, Markovic SN, Nowakowski GS, Ansell SM, Wang ML. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol. 2019;5:1008-1019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 497]  [Article Influence: 99.4]  [Reference Citation Analysis (0)]
35.  Wolmark N, Wieand HS, Rockette HE, Fisher B, Glass A, Lawrence W, Lerner H, Cruz AB, Volk H, Shibata H. The prognostic significance of tumor location and bowel obstruction in Dukes B and C colorectal cancer. Findings from the NSABP clinical trials. Ann Surg. 1983;198:743-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 125]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
36.  Chen HS, Sheen-Chen SM. Obstruction and perforation in colorectal adenocarcinoma: an analysis of prognosis and current trends. Surgery. 2000;127:370-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 147]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
37.  Wang Q, Gao J, Wu X. Pseudoprogression and hyperprogression after checkpoint blockade. Int Immunopharmacol. 2018;58:125-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 77]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
38.  Cohen R, Hain E, Buhard O, Guilloux A, Bardier A, Kaci R, Bertheau P, Renaud F, Bibeau F, Fléjou JF, André T, Svrcek M, Duval A. Association of Primary Resistance to Immune Checkpoint Inhibitors in Metastatic Colorectal Cancer With Misdiagnosis of Microsatellite Instability or Mismatch Repair Deficiency Status. JAMA Oncol. 2019;5:551-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 155]  [Article Influence: 38.8]  [Reference Citation Analysis (0)]
39.  Amodio V, Mauri G, Reilly NM, Sartore-Bianchi A, Siena S, Bardelli A, Germano G. Mechanisms of Immune Escape and Resistance to Checkpoint Inhibitor Therapies in Mismatch Repair Deficient Metastatic Colorectal Cancers. Cancers (Basel). 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
40.  Sahin IH, Akce M, Alese O, Shaib W, Lesinski GB, El-Rayes B, Wu C. Immune checkpoint inhibitors for the treatment of MSI-H/MMR-D colorectal cancer and a perspective on resistance mechanisms. Br J Cancer. 2019;121:809-818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 216]  [Article Influence: 43.2]  [Reference Citation Analysis (0)]
41.  Wang Z, Zhang Q, Qi C, Bai Y, Zhao F, Chen H, Li Z, Wang X, Chen M, Gong J, Peng Z, Zhang X, Cai J, Chen S, Zhao X, Shen L, Li J. Combination of AKT1 and CDH1 mutations predicts primary resistance to immunotherapy in dMMR/MSI-H gastrointestinal cancer. J Immunother Cancer. 2022;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 11]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]