Acessibilidade / Reportar erro

PD-1 blockade as a future treatment for colorectal cancer with microsatellite instability

Bloqueio do PD-1 como futuro tratamento do cancro colorretal com instabilidade de microssatélites

ABSTRACT

Introduction:

Colorectal cancer is the third most common cancer worldwide, with about 15% of these tumours related with microsatellite instability, which confers distinct characteristics to these tumours, both clinicopathological and in the response to treatments. In fact, the poor response to chemotherapy in these tumours has led to the investigation for new treatments, with immunotherapy being the most successful one to date. The focus of this review is to assess the response of microsatellite unstable colorectal cancer to PD-1 blockade, and the mechanisms behind that response.

Methods:

A PubMed research was conducted, resulting in the inclusion of 47 articles in this review.

Results:

Microsatellite instability results in a high neoantigen load, leading to a highly active immune microenvironment of the tumour, mainly due to T-cells. To counteract this, there is an upregulation of PD-1, acting as a “brake” for immune cells, facilitating tumour growth and metastasis. This upregulation makes these tumours great candidates for treatment with PD-1 blockade, as seen in many clinical trials, where the overall responses and progression free survival rates were higher than those observed in microsatellite stable tumours.

Conclusion:

With the importance of colorectal cancer with microsatellite instability new treatments are necessary. Therefore, PD-1 blockade is a promising treatment for colorectal cancer with microsatellite instability, with improvement in survival rates and a better prognosis for these patients.

Keywords:
Colorectal cancer; Microsatellite instability; Immunotherapy; PD-1 receptor; Treatment

Palavras-chave:
Câncer colorretal; Instabilidade de microssatélites; Imunoterapia; Receptor PD-1; Tratamento

RESUMO

Introdução:

O câncer colorretal é o terceiro mais comum em todo o mundo, com cerca de 15% desses tumores relacionados com instabilidade dos microssatélites, o que confere características distintas a esses tumores, tanto clínico patológicas quanto na resposta aos tratamentos. De fato, a fraca resposta à quimioterapia nesses tumores levou à investigação de novos tratamentos, sendo a imunoterapia a mais bem sucedida até o momento. O foco desta revisão é avaliar a resposta do câncer colorretal com microssatélites instáveis ao bloqueio do PD-1 e os mecanismos por trás dessa resposta.

Métodos:

Foi realizada uma pesquisa na base de dados PubMed, resultando na inclusão de 47 artigos nesta revisão.

Resultados:

A instabilidade de microssatélites resulta em uma alta carga de neoantígenos, levando a um microambiente imunológico altamente ativo do tumor, principalmente devido às células T. Para neutralizar isso, há uma maior expressão do PD-1, atuando como um “freio” para as células imunes, facilitando o crescimento do tumor e suas metástases. Essa expressão faz desses tumores grandes candidatos ao tratamento com bloqueio PD-1, como demonstrado em vários ensaios clínicos, onde as respostas globais e as taxas de sobrevivência livres de progressão foram maiores do que as observadas em tumores com microssatélites estáveis.

Conclusão:

Com a importância do câncer colorretal com instabilidade de microssatélites, novos tratamentos são necessários. Portanto, o bloqueio do PD-1 é um tratamento promissor para o câncer colorretal com instabilidade de microssatélites, com melhora nas taxas de sobrevivência e melhor prognóstico para esses pacientes.

Introduction

Colorectal Cancer (CRC) is currently the third most common cancer in men worldwide, and the second in women, with circa 55% occurring in more developed regions, with surgery and chemotherapy remaining the standard of care at the moment.11 Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front. Oncol. 2019;9:396.

In the USA, the 5-year survival rates after surgical removal of CRC for localized (Stage I), regional (Stages II and III) and distant (Stage IV) tumours are 91%, 71% and 13%, respectively.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.

Acquired Somatic Microsatellite Instability (MSI) is seen in 15% of sporadic CRCs, and hereditary MSI is associated with 5% of all CRCs13 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13. but with only 5% of metastatic cancers.55 Colle R, Cohen R, Cochereau D, Duval A, Lascols O, Lopez-Trabada D, et al. Immunotherapy and patients treated for cancer with microsatellite instability. Bull Cancer. 2017;104:42-51.

MSI is associated with dMMR (deficient Mismatch Repair), commonly associated with a better prognosis and improved Overall Survival (OS), except in head and neck cancer and pancreatic cancer.16 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.,77 Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26. Loss of function in one of the mismatch repair proteins leads to accumulation of mistakes in microsatellite regions in the DNA, therefore resulting in MSI and accumulation of replication errors, responsible for about 15% of all CRCs.77 Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26. MSI can be determined by immunohistochemistry, PCR and next-generation sequencing.55 Colle R, Cohen R, Cochereau D, Duval A, Lascols O, Lopez-Trabada D, et al. Immunotherapy and patients treated for cancer with microsatellite instability. Bull Cancer. 2017;104:42-51.

The existing therapies for CRC include surgery for early staged tumours and surgery, chemotherapy and/or radiotherapy in more advanced stages.88 Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019;110:312-8. However, MSI status can be considered a predictive factor of non-response to chemotherapy based in 5-Fluorouracil (5-FU).99 Gupta R, Sinha S, Paul RN. The impact of microsatellite stability status in colorectal cancer. Curr. Probl. Cancer. 2018;42:548-59. It is also known that stage II-III MSI CRCs achieve similar Progression-Free Survival (PFS) and OS, with or without 5-FU-based neoadjuvant chemotherapy,14 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.,66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54. making them good candidates for a different therapeutic approach, namely immunotherapy.33 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,1010 Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019;30:1096-03.1212 Cohen R, Pellat A, Boussion H, Svrcek M, Lopez-Trabada D, Trouilloud I, et al. Immunotherapy and metastatic colorectal cancers with microsatellite instability or mismatch repair deficiency. Bull Cancer. 2019;106:137-42. In fact, after failure of therapy with FOLFOX and FOLFIRI, prolonged survival isn’t common.1313 Fabrizio DA, George TJJr, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J. Gastrointest. Oncol. 2018;9:610-7.

Indeed, tumours with dMMR/MSI are commonly resistant to therapies with methylating agents, platinum compounds and fluoropyrimidines.77 Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26.,1414 Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194.,1515 Lee V, Murphy A, Le DT, Diaz LA. Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist. 2016;21:1200-11.

On the other hand, the improved prognosis of MSI CRCs may be a result of the high anti-tumour immune response of the host.44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13. In fact, tumours with MSI have an important lymphocytic infiltration, making them good candidates for therapies based in the activation of the immune system.77 Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26.

Microsatellite unstable tumours are characteristically tumours with a highly active tumour microenvironment, with dense T-cell infiltrations and upregulation on checkpoint regulators, assuring they are great candidates to Immune Checkpoint Inhibitors (ICIs).1616 Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett. 2019;458:123-35. Besides, MSI is accompanied by a high tumour mutational burden, yet another marker related to the response to ICIs.1717 Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol. Res. 2019;7:1570-3.

Tumours can evade immune surveillance by expressing multiple molecules responsible for the immune response against the tumour, such as Il-10, Transforming Growth Factor-β (TGF-β), immune co-inhibitory signalling proteins such as Programmed cell Death protein 1 (PD-1), Programmed cell Death 1 ligand 1 (PD-L1) and Cytotoxic T Lymphocyte-Associated antigen 4 (CTLA4).1818 Kong P, Wang J, Song Z, Liu S, He W, Jiang C, et al. Circulating lymphocytes, PD-L1 expression on tumor-infiltrating lymphocytes, and survival of colorectal cancer patients with different mismatch repair gene status. J. Cancer. 2019;10:1745-54. PD-1 acts as a “brake” for immune cells inhibiting T-cells, and therefore the immune response to tumours.116 Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett. 2019;458:123-35.,1919 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74. Thus, the PD-1/PD-L1 axis may be responsible for inducing immune inhibition or exhaustion, resulting in a decrease in activated T-cells. Therefore, the anti-tumour immune response is impaired, making this the rationale for the treatment with blockade of this axis, which allows the recovering of native anti-tumour function of T-cells, facilitating tumour regression.2020 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6. Inhibitors of the PD1/PD-L1 axis interrupt the immunoediting incited by this axis, resulting in stimulation of T cells and antitumor activity.1515 Lee V, Murphy A, Le DT, Diaz LA. Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist. 2016;21:1200-11.

With the increase in OS of patients with CRC, there is also an increase in the cost of treatment,2121 Messersmith WA. NCCN Guidelines Updates: management of metastatic colorectal cancer. J. Natl. Compr. Canc. Netw. 2019;17:599-601. making the choice of treatment even more important.

With the favourable outcomes of these immunotherapies, the Food and Drug Administration (FDA) approved pembrolizumab for the treatment of adult and paediatric patients with advanced MSI/dMMR tumours in the 23rd of May 2017. Furthermore, on the 1st of July 2017, an accelerated approval was granted by the FDA for the use of nivolumab in patients with metastatic CRC with a MSI/dMMR phenotype.2222 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17. Besides, FDA has also approved dMMR/MSI as indicators of response to ICIs for metastatic cancers irrespective of cancer type.66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.

This review is aimed at understanding the characteristics of CRC with MSI and the ways in which MSI influences tumour response to immunotherapy, focusing on the role of PD-1 blockade in the treatment of CRC with MSI.

Methods

In this narrative review, a web search was done on the PubMed database on September 2019, using the following query: “(((Immunotherapy OR pembrolizumab OR nivolumab)) AND Colorectal Cancer) AND (Microsatellite instability OR MSI)”. Using this query, a total of 245 articles were obtained.

When refining the search for articles published only in the last five years and filtering for the languages of Portuguese and English, a total of 226 articles were obtained. No exclusion criteria were used (Fig. 1). Based on the title, 97 articles were excluded mainly due to the referral of other tumour types such as melanoma, endometrium and urological. This exclusion was also based on studies based only on specific populations, genetic alterations and on genomic sequencing. With this exclusion, 129 articles were analysed based on their abstract. This analysis resulted in the exclusion of 34 articles, with articles focusing on tumour diagnosis methods, explanation of tumour pathophysiology, studies with the main focus on microsatellite stable tumours and only specific populations studied.

Fig. 1
Methods.

This resulted in a total of 95 articles remaining for the full text analysis, with the exclusion of 48 articles, mainly due to the unavailability of the full text and the language of the article (some articles in Czech and French). Besides, some articles were also excluded because of their outdated information (mainly the articles from 2015). With this process, 47 articles were selected and included in this review.

MeSH terms

“immunotherapy”; “microsatellite instability”; “nivolumab”; “colorectal neoplasms”

Supplementary Concept: “pembrolizumab”.

Characteristics of CRC with MSI

MSI manifests with an increased variability in the size of microsatellites throughout the patients´ genome, leading to further DNA mutations, which increases neoantigen production, making the tumour more recognizable to the immune system.44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.,66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.,2323 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12. Associated with the increase in dendritic cells, this facilitates the presentation of tumour cells to cytotoxic T lymphocytes, resulting in an increase in tumour infiltrating lymphocytes, mainly CD8+. This is, in part, related to the better prognosis usually associated with CRC with MSI.44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.

5 Colle R, Cohen R, Cochereau D, Duval A, Lascols O, Lopez-Trabada D, et al. Immunotherapy and patients treated for cancer with microsatellite instability. Bull Cancer. 2017;104:42-51.

6 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.

7 Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26.

8 Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019;110:312-8.

9 Gupta R, Sinha S, Paul RN. The impact of microsatellite stability status in colorectal cancer. Curr. Probl. Cancer. 2018;42:548-59.

10 Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019;30:1096-03.

11 Miyauchi T, Yaguchi T, Kawakami Y. Inter-patient and intra-tumor heterogeneity in the sensitivity to tumor-targeted immunity in colorectal cancer. Nihon Rinsho. Meneki. Gakkai. Kaishi. 2017;40:54-9.

12 Cohen R, Pellat A, Boussion H, Svrcek M, Lopez-Trabada D, Trouilloud I, et al. Immunotherapy and metastatic colorectal cancers with microsatellite instability or mismatch repair deficiency. Bull Cancer. 2019;106:137-42.

13 Fabrizio DA, George TJJr, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J. Gastrointest. Oncol. 2018;9:610-7.

14 Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194.

15 Lee V, Murphy A, Le DT, Diaz LA. Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist. 2016;21:1200-11.

16 Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett. 2019;458:123-35.

17 Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol. Res. 2019;7:1570-3.

18 Kong P, Wang J, Song Z, Liu S, He W, Jiang C, et al. Circulating lymphocytes, PD-L1 expression on tumor-infiltrating lymphocytes, and survival of colorectal cancer patients with different mismatch repair gene status. J. Cancer. 2019;10:1745-54.

19 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74.

20 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6.

21 Messersmith WA. NCCN Guidelines Updates: management of metastatic colorectal cancer. J. Natl. Compr. Canc. Netw. 2019;17:599-601.

22 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17.

23 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12.
-2424 Quiroga D, Lyerly HK, Morse MA. Deficient mismatch repair and the role of immunotherapy in metastatic colorectal cancer. Curr. Treat. Options Oncol. 2016;17:41.

MSI CRC is clinicopathologically different than other subtypes of CRC, namely with a younger age of onset, proximal location, lymphocytic reaction, mucinous/signet ring differentiation and a medullary growth pattern. Besides, MSI is usually associated with a reduced risk for nodal and distant metastases, with an increased Disease Free Survival (DFS) in early stages.14 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.,66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.

The majority of MSI CRCs are included in the Molecular Subtype 1 of the Consensus Molecular Subtype Classification, a group of hypermutated tumours with strong immune activation.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.,33 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,2525 Kather JN, Halama N, Jaeger D. Genomics and emerging biomarkers for immunotherapy of colorectal cancer. Semin. Cancer Biol. 2018;52:189-97. This subgroup is usually associated with a better prognosis, with intratumoral heterogeneity (mixed conventional, mucinous, and poorly differentiated carcinoma), and a prominent inflammatory reaction at the advancing edge of the tumour (Crohn-like reaction). Tumour infiltrating lymphocytes are mainly CD8+ cytotoxic T-cells.33 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.

MSI is associated with a high mutation load, with MSI tumours presenting with a mutational rate 20 times higher than tumours with Microsatellite Stability (MSS).2222 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17. This high tumour mutational load is also typically seen in right-sided CRC.2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34. MSI CRC are associated with a higher tumour mutational load, especially with high Microsatellite Instability (MSI-H), and a high tumour mutational load is in turn associated with MSI-H (p < 0.0001).2323 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12.

In fact, MSI is essentially a marker of dMMR, which in turn results in an inhability to correct possible DNA mistakes that arise during replication, therefore increasing the tumour mutational load.16 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.,2222 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17. The higher mutational burden results in neoantigens, triggering the host immune system and increasing T-cell infiltration127 Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019;16:361-75.,2828 Lee KS, Kwak Y, Ahn S, Shin E, Oh HK, Kim DW, et al. Prognostic implication of CD274 (PD-L1) protein expression in tumor-infiltrating immune cells for microsatellite unstable and stable colorectal cancer. Cancer Immunol. Immunother. 2017;66:927-39. and, therefore, the tumour immunogenicity. These tumours have a higher activation of CD8+ T-cells, as well as an upregulation of checkpoints such as CTLA-4 and PD-1/PD-L1, amongst others.11 Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front. Oncol. 2019;9:396.,2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34. However, tumour cells are not eliminated by the immune system because of the upregulation of inhibitory checkpoints (such as PD-1, CTLA4, LAG3, and Indoleamine 2,3-Dioxygenase)(IDO),2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34. which in turn favour an immune-suppressive microenvironment.2828 Lee KS, Kwak Y, Ahn S, Shin E, Oh HK, Kim DW, et al. Prognostic implication of CD274 (PD-L1) protein expression in tumor-infiltrating immune cells for microsatellite unstable and stable colorectal cancer. Cancer Immunol. Immunother. 2017;66:927-39.

Microsatellite unstable CRCs are associated with a high expression of PD-1/PD-L1. This signalling axis may be associated with immune inhibitory/exhaustion signalling to activated T-cells resulting in a significant impairment of antitumour immune response.2020 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6. PD-L1 positivity in MSI CRCs immune cells was significantly correlated with an early stage disease (81%), peritumoural lymphoid reaction (77%) and a high density of tumour infiltrating lymphocytes (77%), whereas PD-L1 positivity in tumour cells was associated with a more advanced disease stage (73%), lymphovascular invasion (46%), high density of tumour infiltrating lymphocytes (81%) and a poorer differentiation (65%).2020 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6. It was observed that tumours with higher positivity for PD-L1 were associated with high tumour infiltrating lymphocytes and peritumoural infiltration, which in turn is associated with a better prognosis.2020 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6.

MSI tumours are associated with tumour infiltrating immune cells that are PD-L1 positive, but not with tumour cells expressing this marker.2929 Ho HL, Chou TY, Yang SH, Jiang JK, Chen WS, Chao Y, et al. PD-L1 is a double-edged sword in colorectal cancer: the prognostic value of PD-L1 depends on the cell type expressing PD-L1. J. Cancer Res. Clin. Oncol. 2019;145:1785-94. The expression of PD-L1 in stromal tumour infiltrating immune cells is associated with a good prognosis (p < 0.001), however the presence of tumour cells expressing PD-L1 is a marker of poor prognosis. MSI-H CRC is associated with the presence of stromal tumour infiltrating immune cells expressing PD-L1 but not tumour cells expressing PD-L1.2929 Ho HL, Chou TY, Yang SH, Jiang JK, Chen WS, Chao Y, et al. PD-L1 is a double-edged sword in colorectal cancer: the prognostic value of PD-L1 depends on the cell type expressing PD-L1. J. Cancer Res. Clin. Oncol. 2019;145:1785-94. dMMR patients show higher tumour infiltrating lymphocytes PD-1 (54.8% vs. 43.2%, p = 0.039) and lower stroma PD-L1 (non-tumour infiltrating lymphocyte and non-tumour) expression (22.0% vs. 43.8%, p < 0.001), when compared to proficient Mismatch Repair (pMMR) patients.1818 Kong P, Wang J, Song Z, Liu S, He W, Jiang C, et al. Circulating lymphocytes, PD-L1 expression on tumor-infiltrating lymphocytes, and survival of colorectal cancer patients with different mismatch repair gene status. J. Cancer. 2019;10:1745-54.

The higher density of CD8+ T-cells is associated with CRC tumours that don’t have early metastases and with prolonged OS. In fact, high risk early stage CRCs have a low CD8+ T-cells density.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47. Besides, MSI is associated with a positive prognosis in the early stages of CRC, without adjuvant chemotherapy, having the opposite effect and a worse prognosis with FU-based chemotherapy.66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.

Tumour microenvironment in microsatellite unstable CRC

The tumour microenvironment is made of tumour-associated antigens, antigen-presenting cells, T lymphocytes (that recognize and destroy cells marked with tumour-associated antigens), B lymphocytes (producing antibodies to tumour antigens) and stromal cells.2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34. In order to get an immune response to tumour cells, a reactivation of T-cells in the tumour microenvironment is required.1919 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74. tumour microenvironment with higher density of CD8+ tumour infiltrating lymphocytes are accompanied by high numbers of IFN and IFN-related cytokines, making it the most responsive to immunotherapy.1919 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74.

Tumour infiltrating lymphocytes are a mixture of T and B lymphocytes, NK cells, macrophages and other innate cells. MSI is associated with a high number of tumour infiltrating lymphocytes and lymphoid reaction, which is a good prognosis factor.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47. Tumours with high levels of tumour infiltrating lymphocytes are tumours located to the proximal colon, with H-MSI and high neoantigen load (p < 0.001).3030 Hamad T, Soong TR, Masugi Y, Kosumi K, Nowak JA, da Silva A, et al. TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PD-L1) expression status and tumor-infiltrating lymphocytes in colorectal carcinomas. Oncoimmunology. 2018;7:e1442999.

MSI-H CRCs usually have a prominent inflammatory response in the edge of the tumour, with an increased number of tumour infiltrating lymphocytes, mainly T cytotoxic cells (CD8+).13 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,3131 Maby P, Tougeron D, Hamieh M, Mlecnik B, Kora H, Bindea G, et al. Correlation between Density of CD8+ T-cell infiltrate in microsatellite unstable colorectal cancers and frameshift mutations: a rationale for personalized immunotherapy. Cancer Res. 2015;75:3446-55. This pro-inflammatory status in MSI is associated with less lymphovascular invasion and metastasis, related to the better prognosis.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.,3030 Hamad T, Soong TR, Masugi Y, Kosumi K, Nowak JA, da Silva A, et al. TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PD-L1) expression status and tumor-infiltrating lymphocytes in colorectal carcinomas. Oncoimmunology. 2018;7:e1442999.,3232 Riley JM, Cross AW, Paulos CM, Rubinstein MP, Wrangle J, Camp ER. The clinical implications of immunogenomics in colorectal cancer: A path for precision medicine. Cancer. 2018;124:1650-9.

A lower density of CD4+ and CD8+ T-cells is, in turn, associated with primary CRC with higher metastatic potential. Furthermore, the higher density of myeloid-derived suppressor cells and Th17 cells is significantly associated with tumour progression and a poorer prognosis of CRC, related to the immune suppressive effects of these cells.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.

The immunoscore quantifies the number, density, and distribution of CD3+ T lymphocytes and CD8+ cytotoxic T-cells in the tumour core and its invasive margins and may even provide a good staging tool given its prognostic value. Tumours with high immunoscore (3 and 4) are associated with a better prognosis.13 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,3131 Maby P, Tougeron D, Hamieh M, Mlecnik B, Kora H, Bindea G, et al. Correlation between Density of CD8+ T-cell infiltrate in microsatellite unstable colorectal cancers and frameshift mutations: a rationale for personalized immunotherapy. Cancer Res. 2015;75:3446-55. In fact, in tumours with low immunoscore, 72% of these relapsed after 5 years, versus 4.7% of patients with a high immunoscore.3232 Riley JM, Cross AW, Paulos CM, Rubinstein MP, Wrangle J, Camp ER. The clinical implications of immunogenomics in colorectal cancer: A path for precision medicine. Cancer. 2018;124:1650-9. The immune infiltration in the tumour is inversely associated with early metastasis and death.130 Hamad T, Soong TR, Masugi Y, Kosumi K, Nowak JA, da Silva A, et al. TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PD-L1) expression status and tumor-infiltrating lymphocytes in colorectal carcinomas. Oncoimmunology. 2018;7:e1442999.,3232 Riley JM, Cross AW, Paulos CM, Rubinstein MP, Wrangle J, Camp ER. The clinical implications of immunogenomics in colorectal cancer: A path for precision medicine. Cancer. 2018;124:1650-9.

Immunoediting is a process that occurs when the immune system attempts to eliminate tumour cells, following the designated elimination, equilibrium, and escape sequence. This process is regulated by checkpoint receptors mainly CTLA-4, PD-1/PD-L1 and LAG3.2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34. PD-1/PD-L1 checkpoint blocks the interaction of tumour-expressed PD-L1 and PD-1 expressed in T-cells, inactivating T-cells and allowing the tumour to grow.2626 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34.

The presence of Treg cells in tumour infiltrating lymphocytes produce immunosuppressive substrates (like IL-10) and express several immune checkpoints namely PD-L1 and CTLA-4, contributing to the inhibition of effector T-cells. A tumour microenvironment enriched with CD8+ is associated with a reduced recurrence and metastasis, whereas a tumour microenvironment enriched with immunosuppressive cancer associated fibroblasts is associated with a poorer prognosis.22 Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.

MSI CRC is associated with PD-L1 expression in tumour infiltrating immune cells, both stromal (p = 0.042) and intraepithelial (p < 0.001).2929 Ho HL, Chou TY, Yang SH, Jiang JK, Chen WS, Chao Y, et al. PD-L1 is a double-edged sword in colorectal cancer: the prognostic value of PD-L1 depends on the cell type expressing PD-L1. J. Cancer Res. Clin. Oncol. 2019;145:1785-94. The presence of these stromal cells was a marker of good prognosis (p < 0.001), whereas the presence of tumour cells expressing PD-L1 was a marker of poor prognosis.2929 Ho HL, Chou TY, Yang SH, Jiang JK, Chen WS, Chao Y, et al. PD-L1 is a double-edged sword in colorectal cancer: the prognostic value of PD-L1 depends on the cell type expressing PD-L1. J. Cancer Res. Clin. Oncol. 2019;145:1785-94. Expression of PD-L1 in tumour infiltrating lymphocytes of patients with microsatellite unstable CRC is 68.6%, higher than the observed in MSS CRC.66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.

Large mucinous areas surrounding the tumour possibly result from immune destruction of tumour cells, making extracellular mucin a type of treatment response marker.3333 Llosa NJ, Luber B, Siegel N, Awan AH, Oke T, Zhu Q, et al. Immunopathologic stratification of colorectal cancer for checkpoint blockade immunotherapy. Cancer Immunol. Res. 2019;7:1574-9. The CRC who presented clinical benefit from ICIs tend to present with high PD-L1 expression and a mucinous pattern.3333 Llosa NJ, Luber B, Siegel N, Awan AH, Oke T, Zhu Q, et al. Immunopathologic stratification of colorectal cancer for checkpoint blockade immunotherapy. Cancer Immunol. Res. 2019;7:1574-9.

Microsatellite testing in patients with CRC

The current gold standard for assessing tumour Mismatch Repair (MMR) activity is molecular testing for MSI. It is also possible to assess MMR by immunohistochemistry (to identify the loss of one or more of the MMR proteins: MLH1, MSH2, MSH6 and PMS2), which is sensitive but not specific.13 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.

In sporadic MSI CRCs, about 95% of the cases are associated with MLH1 promoter hypermethylation, with loss of function of MLH1 protein.13 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.,66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54. The reported sensitivity of DNA testing for MSI is 89% for MLH1/MSH2 and 77% for MSH6.44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.

If 30% or more of the repeats tested on PCR are unstable, the tumour is classified as MSI-H.14 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.,66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54. MSI testing with either method is found to be cost-effective, sensitive and specific, with benefits widely accepted.44 Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.

Furthermore, if the patient is shown to have MSI, they should be given the option of genetic counselling, to determine the germline mutation in the MMR gene.33 Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.

MSI is predictive of tumour response to conventional chemotherapy and immunotherapy, with consequences on the prognosis.66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54. MSI is a response biomarker for PD-1 blockade, with PFS rates of as much as 78% in MSI-H CRC patients, versus only 11% of MSS patients.1313 Fabrizio DA, George TJJr, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J. Gastrointest. Oncol. 2018;9:610-7.

MSI is associated with favourable outcomes when compared with MSS tumours, a reason why some American guidelines recommend that MSI should be tested upon the CRC diagnosis.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. With these results, MSI testing in patients with CRC can improve the prognosis is this subset of patients.

MSI as a biomarker of response to immunotherapy

MSI-H results in a high number of DNA replication errors that can be recognized as foreign by the host immune cells, eliciting an enhanced immune infiltration, making MSI a biomarker for the response to ICIs.3535 Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41. The high infiltration of CD8+ T cells in MSI tumours ensures a more durable response to immunotherapy.66 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54. This was shown in the KEYNOTE-016 trial, where a much higher Overall Response Rate (ORR) was observed in MSI tumours when compared to MSS tumours (40% vs. 0%, respectively).3535 Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41.

MSI is a marker of dysfunction in MMR proteins, giving rise to an array of DNA mutations and neoantigens, with this higher tumour mutational burden as a marker of response to ICIs.2222 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17. Cases with ORR to immunotherapies tend to have a higher tumour mutational burden (54 mutations/Mb) when compared to non-responders (29 mutations/Mb) (p < 0.001).1010 Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019;30:1096-03. Therefore dMMR genotype is considered a probable marker of response to PD-1 inhibitors.16 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.,88 Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019;110:312-8.

Besides MSI, PD-L1 protein expression is also associated with benefit from immunotherapy.1414 Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194. MSI is associated with a higher expression of PD-1/PD-L1 when compared to MSS (20.6% vs. 7.8%, p < 0.0001),2323 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12. associated with a good response to anti-PD1/PD-L1 therapies.2020 Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6. However, assessment of PD-L1 expression and tumour mutational burden is costly and not as easily accessible as MSI testing, making MSI a more feasible biomarker of response to immunotherapy.1414 Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194.

In summary, to balance the active tumour microenvironment in MSI CRC, tumour cells express immune checkpoint ligands, promoting an immune-suppressive environment. This is the rationale for why MSI is a good predictive biomarker of response to checkpoint inhibitors.1414 Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194.,1818 Kong P, Wang J, Song Z, Liu S, He W, Jiang C, et al. Circulating lymphocytes, PD-L1 expression on tumor-infiltrating lymphocytes, and survival of colorectal cancer patients with different mismatch repair gene status. J. Cancer. 2019;10:1745-54.,2323 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12.,2828 Lee KS, Kwak Y, Ahn S, Shin E, Oh HK, Kim DW, et al. Prognostic implication of CD274 (PD-L1) protein expression in tumor-infiltrating immune cells for microsatellite unstable and stable colorectal cancer. Cancer Immunol. Immunother. 2017;66:927-39.,3232 Riley JM, Cross AW, Paulos CM, Rubinstein MP, Wrangle J, Camp ER. The clinical implications of immunogenomics in colorectal cancer: A path for precision medicine. Cancer. 2018;124:1650-9. Furthermore, it is known that MSI, tumour mutational load and PD-1/PD-L1 expression are all markers associated with response to ICIs.2323 Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12.

Response of microsatellite unstable CRC to immunotherapy

Several studies have been conducted in order to sustain the benefits of immunotherapy in MSI CRC (Table 1). Nivolumab is a human IgG4 monoclonal antibody against PD-1, while pembrolizumab is a humanized IgG4 monoclonal antibody that binds to PD-1, preventing the interactions in the PD-1/PD-L1 axis. Therefore, both antibodies result in immune recognition of the tumour and subsequent response.3636 Arora SP, Mahalingam D. Immunotherapy in colorectal cancer: for the select few or all?. J. Gastrointest. Oncol. 2018;9:170-9. The first-line therapy is key as it can determine the successful systemic treatment in metastatic CRC (since it has the longest duration). However, pembrolizumab and nivolumab are currently only second-line and beyond for patients with MSI/Dmmr.16 Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.,3737 Modest DP, Pant S, Sartore-Bianchi A. Treatment sequencing in metastatic colorectal cancer. Eur. J. Cancer. 2019;109:70-83.

Table 1
Main studies and results regarding PD-1 blockade in CRC.

The Checkmate-142 study was done in order to investigate the activity and safety of nivolumab in patients with locally determined dMMR/MSI metastatic or recurrent CRCs. Seventy-four patients were recruited and enrolled in this study, receiving 3 mg/kg intravenous nivolumab monotherapy every 2 weeks until disease progression, death, unacceptable toxic effects, withdrawal of consent or ending of the study occurred.3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91. Besides tumour assessment and MSI/dMMR testing, the level of expression of PD-1 was also measured. The median follow-up was 12 months.3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91. Twenty-three (31.1%) patients achieved an objective response and disease control for 12 weeks or longer occurred in 51 patients (69%). These responses seemed durable as only three responders experienced progression.3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91. The median PFS was 14.3 months (with 50% PFS at 12 months).3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91. Twelve-month OS was 73% with clinically important improvements in function, symptoms and global quality of life manifesting as early as week 13 of treatment. These outcomes were maintained for more than 37 weeks.3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91.

Posteriorly, a second phase of the Checkmate-142 study was conducted in order to assess the benefit of adding anti-CTLA4 therapy (ipilimumab) to anti-PD1 therapy (nivolumab). One-hundred and nineteen patients with histologically confirmed recurrent or metastatic CRC assessed as dMMR and/or MSI-H received nivolumab 3 mg/kg + ipilimumab 1 mg/kg once every 3 weeks for four doses followed by nivolumab 3 mg/kg IV once every 2 weeks.3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9. Fourty-four patients discontinued therapy due to disease progression (19%), adverse events related to the treatment (13%) and adverse events unrelated to the drug in study (2%). An objective response was achieved in 54.6% of patients (3.4% Complete Response [CR] and 51.3% partial response). In 80% of patients, disease control for ≥ 12 weeks was achieved. The ORR was 49% (4% CR and 45% partial response). Furthermore, the response was maintained, with 94% of responders having ongoing responses at data cut off and 83% lasting longer than 6 months.3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9. Furthermore, improvements in functioning, global health status and symptoms were clinically important and statistically significant. These results favour a hypothesis that combination therapy with ICIs is beneficial.3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9.

Another clinical trial (KEYNOTE-016) aimed to assess the response of pMMR and dMMR chemo-refractory metastatic CRC or non-CRCs with dMMR to the anti-PD1 monoclonal antibody pembrolizumab. In the beginning of this trial 41 patients were included and treated with 10 mg/kg of pembrolizumab every 3 weeks. Ten participants were dMMR CRC, with an ORR of 40% and PFS rate of 78%. In the updated trial, 86 patients with dMMR cancers were included, with an ORR of 52% (12% CR in the CRC cohort).135 Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41.,4040 Ciardiello D, Vitiello PP, Cardone C, Martini G, Troiani T, Martinelli E, et al. Immunotherapy of colorectal cancer: Challenges for therapeutic efficacy. Cancer Treat. Rev. 2019;76:22-32. The PFS rate was 59% at two years, and the OS rate at two years was 72%.4040 Ciardiello D, Vitiello PP, Cardone C, Martini G, Troiani T, Martinelli E, et al. Immunotherapy of colorectal cancer: Challenges for therapeutic efficacy. Cancer Treat. Rev. 2019;76:22-32.

The Keynote-164 trial evaluated the response of MSI/dMMR CRC previously treated with at least two therapies including fluoropyrimidine, oxaliplatin, and irinotecan (Cohort A, with 61 patients) and in patients previously treated with at least one prior therapy including fluoropyrimidine, oxaliplatin, irinotecan, or anti- VEGF/EGFR (Cohort B, with 63 patients).3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. In Cohort A the ORR was 28% with a PFS at 12 months of 34%, and an OS of 72% at twelve months. On the other hand, in Cohort B, the ORR was 32%, with a 12-month PFS of 41% and an OS of 76% at twelve months. These results favour the utilization of pembrolizumab given its efficacy.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. In this trial, the CR was 3%, higher than the usual 1% using combination chemotherapy, similar to the one observed in the CheckMate-142 trial, where the CR rose to 9% when the follow-up was prolonged to 21 months, meaning the response to treatment increases with time.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45.

Keynote-028 was a multicentre, open-label, nonrandomized, single-arm phase Ib trial with 23 patients with metastatic or locally advanced colon or rectal adenocarcinoma. Patients received pembrolizumab intravenously at a dose of 10 mg/kg once every 2 weeks for 24 months (or until confirmed disease progression).4141 O’Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One. 2017;12:e0189848. In this trial 1 patient experienced partial response and 17% of patients remained with stable disease, with a median duration of 5.1 months. Among the remaining patients, 65% had progressive disease. The ORR was 13%. The one patient that responded to pembrolizumab was the only one in this cohort of patients with MSI CRC, stating the importance of this biomarker to the response to PD-1 blockade.4141 O’Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One. 2017;12:e0189848.

Keynote-177 is a future clinical trial proposed to study the comparison between standard chemotherapy and pembrolizumab in 270 previously untreated dMRR/MSI-H metastatic CRC, unlike the other existing trials, where the patients have previously been treated.4242 Stein A, Folprecht G. Immunotherapy of colon cancer. Oncol. Res. Treat. 2018;41:282-5.

Notably, primary resistance to ICIs vary from 12% to 40% of patients, with several mechanisms possibly responsible such as JAK loss-of-function mutations, truncating mutations of beta-2-microglobulin or even loss of Major Histocompatibility Complex (MHC) molecules.112 Cohen R, Pellat A, Boussion H, Svrcek M, Lopez-Trabada D, Trouilloud I, et al. Immunotherapy and metastatic colorectal cancers with microsatellite instability or mismatch repair deficiency. Bull Cancer. 2019;106:137-42.,4343 Yeon Yeon S, Jung SH, Jo YS, Choi EJ, Kim MS, Chung YJ, et al. Immune checkpoint blockade resistance-related B2M hotspot mutations in microsatellite-unstable colorectal carcinoma. Pathol. Res. Pract. 2019;215:209-14. Besides that, weak responses to ICIs are related to low expression of genes associated with infiltration of immune cells, mainly Th1 and cytotoxic T-cells.88 Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019;110:312-8. It is known, however, that tumour response to ICIs was independent of KRAS and BRAF status,4242 Stein A, Folprecht G. Immunotherapy of colon cancer. Oncol. Res. Treat. 2018;41:282-5. known to have implications in the response to other treatments.

Possible adverse effects of treatment with immunotherapy

The most common side effect of ICIs is skin toxicity (rash and pruritus), in about 20% of patients. Less than 10% of side effects include cardiac arrhythmias, infusion-related reactions, iridocyclitis and peripheral and sensory neuropathy. Laboratory findings include disturbances in liver enzymes, hyponatremia, hyperkalaemia, disturbances in thyroid function and in cortisol levels.2222 Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17. Nevertheless, in 71%−96% of patients drug-related adverse events resolved, with the exception of endocrine events, resolving in 40% of patients.3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9.

In fact, about 70% of the patients in the CheckMate-142 trial had drug-related adverse effects. However, serious drug-related adverse events were only observed in 12% of patients, including adrenal insufficiency, ALT levels increase, colitis, gastritis, stomatitis, acute kidney injury and arthritis. In fact, no deaths related to the study drug were reported.3838 Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91.

Combination therapy with nivolumab + ipilimumab results in an increase in adverse effects, with high levels of AST and/or ALT (11%), elevated lipase (4%), anaemia (3%) and colitis (3%) being the most common.112 Cohen R, Pellat A, Boussion H, Svrcek M, Lopez-Trabada D, Trouilloud I, et al. Immunotherapy and metastatic colorectal cancers with microsatellite instability or mismatch repair deficiency. Bull Cancer. 2019;106:137-42.,3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9. A case report showed a 61-year old woman who received nivolumab + ipilimumab developing rapidly extending proximal muscles weakness with significant muscular oedema, enhancing the possibility of synergy in adverse events when combining ICIs.4444 Tauber M, Cohen R, Laly P, Josselin L, Andre T, Mekinian A. Severe necrotizing myositis associated with long term anti-neoplastic efficacy following nivolumab plus ipilimumab combination therapy. Clin. Rheumatol. 2019;38:601-2.

Most adverse events are noted within 3 − 12 weeks of treatment,134 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45.,4545 Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8. resolving within 12 weeks of onset.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. Some of the most common adverse events (skin rash, pruritus) can respond to antihistamines, and topical steroids,4545 Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8. which do not appear to interfere with the therapeutic effect of immunotherapy.1919 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74. If this fails, early addition of immunosuppressive drugs (such as infliximab) may be considered.4545 Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8. Despite having adverse events related to treatment, PD-1/PD-L1 inhibitors have a significantly lower risk of fatigue, neuropathy, diarrhoea, hematologic toxicity, anaemia, nausea and anorexia, when compared with chemotherapy.1919 Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74.

Incorporating immunotherapy onto the treatment of MSI CRC

PD-1 blockers have been associated with good outcomes in the treatment of MSI CRC with durable responses, good survival rates and an acceptable safety profile, making them favourable for the treatment of these patients, especially given their low response rates to chemotherapy.3939 Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9.

In fact, pembrolizumab was approved by the FDA for all solid tumours with dMMR, regardless of histology, after failure of treatment with first-line therapy. Furthermore, nivolumab was also approved by the FDA, but only in MSI CRCs, after disease progression with chemotherapy with fluoropyrimidine, oxalipatin and irinotecan.127 Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019;16:361-75.,4545 Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8. Currently, pembrolizumab and nivolumab are considered second-line treatments for CRC, and should only be used after molecular profiling of the tumour.3737 Modest DP, Pant S, Sartore-Bianchi A. Treatment sequencing in metastatic colorectal cancer. Eur. J. Cancer. 2019;109:70-83.

Nevertheless, therapy with ICIs is expensive (reaching as far as 300,000 $/year per patient regarding ipilimumab + nivolumab combination). It isn’t the most cost-effective, mainly due to the drug pricing in nivolumab. When talking about first-line therapy, the combination therapy was the most effective, however still not cost-effective in comparison with mFOLFOX6 and cetuximab. Nonetheless, the OS with ipilimumab + nivolumab was longer, with patients more likely to avoid chemotherapy, making these therapies a good choice of treatment in CRC with MSI, especially if the pricing of the drugs is lowered.4646 Chu JN, Choi J, Ostvar S, Torchia JA, Reynolds KL, Tramontano A, et al. Cost-effectiveness of immune checkpoint inhibitors for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Cancer. 2019;125:278-89.

For the majority of patients, first-line treatment remains combination chemotherapy with targeted therapy. However, for patients with MSI/dMMR, it is recommended the use of nivolumab (with or without ipilimumab) or pembrolizumab as one of several first-line regimens, especially in patients where chemotherapy cannot be done.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. Importantly, the MSI status should be assessed before treatment, given these ICIs have greater outcomes in this subset of patients.3434 Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45. Indeed, the incorporation of PD-1 blockers in the treatment of CRC is of the greatest interest, given their clinical benefits and safety profiles, especially after their approval by the NCCN.127 Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019;16:361-75.,4545 Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8.

Alternative immunotherapies

Besides ICIs, there are other immunotherapies in study for the treatment of CRC. For example, cancer autologous vaccines use the patient’s tumour cells containing tumour-associated antigens, with limited efficacy to date.3636 Arora SP, Mahalingam D. Immunotherapy in colorectal cancer: for the select few or all?. J. Gastrointest. Oncol. 2018;9:170-9. Another option is adoptive cell transfer, treating patients with cell populations previously expanded ex vivo, in an attempt to enable the patients’ T-cells to overcome the immune suppression occurring in vivo.127 Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019;16:361-75.,3535 Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41. Toll-like receptors agonists are also under investigation.135 Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41.,3636 Arora SP, Mahalingam D. Immunotherapy in colorectal cancer: for the select few or all?. J. Gastrointest. Oncol. 2018;9:170-9. Other novel therapies under investigation include inhibitors of enzymes responsible for generating nutritional stress for T-cells, such as IDO and immunomodulatory molecules like CD73.4747 Rawla P, Barsouk A, Hadjinicolaou AV, Barsouk A. Immunotherapies and targeted therapies in the treatment of metastatic colorectal cancer. Med. Sci. (Basel). 2019;7.

Conclusion

CRC is still one of the most common tumours worldwide, with about 15% of these tumours related with MSI. MSI tumours have distinct characteristics, namely the high neoantigen load and the enhanced immune infiltration. These characteristics have implications not only in the prognosis, but also when it comes to the response to different treatments, consequently justifying MSI testing before the treatment of patients with CRC.

In fact, despite the better prognosis of MSI tumours, their worse response to chemotherapy is known, enhancing the need for other therapies when treating these patients. Moreover, the high T-cell infiltration in these tumours favours the upregulation of checkpoint regulators, where the PD-1/PD-L1 axis has a main role, resulting in an immune system inactivation, thus facilitating tumour expansion and growth.

Similarly, with these characteristics of MSI CRC, it is possible to conclude the important role of immune checkpoint blockade in the treatment of these tumours, namely with the use of anti-PD1 antibodies like pembrolizumab and nivolumab. In fact, studies exploring the effects of PD-1 blockade in the treatment of patients with MSI CRC have demonstrated the high efficacy of the aforementioned antibodies in this context, with an acceptable safety profile.

In conclusion, patients with MSI CRC have tumours that justify a different approach, with PD-1 blockade being a good treatment alternative in these cases. Nonetheless, patients should be tested for MSI before initiating these treatments and approval by the regulatory authorities is required. In short, PD-1 blockade can improve symptoms, ORR and OS in patients with MSI CRC, making these antibodies one of the future treatments of these patients.

References

  • 1
    Oliveira AF, Bretes L, Furtado I. Review of PD-1/PD-L1 Inhibitors in Metastatic dMMR/MSI-H Colorectal Cancer. Front. Oncol. 2019;9:396.
  • 2
    Koi M, Carethers JM. The colorectal cancer immune microenvironment and approach to immunotherapies. Future Oncol. 2017;13:1633-47.
  • 3
    Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part I-Colorectal Cancer: Microsatellite Instability, Testing, and Clinical Implications. Arch Pathol. Lab Med. 2018;142:17-25.
  • 4
    Chang L, Chang M, Chang HM, Chang F. Expending role of microsatellite instability in diagnosis and treatment of colorectal cancers. J. Gastrointest. Cancer. 2017;48:305-13.
  • 5
    Colle R, Cohen R, Cochereau D, Duval A, Lascols O, Lopez-Trabada D, et al. Immunotherapy and patients treated for cancer with microsatellite instability. Bull Cancer. 2017;104:42-51.
  • 6
    Zhao P, Li L, Jiang X, Li Q. Mismatch repair deficiency/microsatellite instability-high as a predictor for anti-PD-1/PD-L1 immunotherapy efficacy. J. Hematol. Oncol. 2019;12:54.
  • 7
    Gelsomino F, Barbolini M, Spallanzani A, Pugliese G, Cascinu S. The evolving role of microsatellite instability in colorectal cancer: a review. Cancer Treat. Rev. 2016;51:19-26.
  • 8
    Yaghoubi N, Soltani A, Ghazvini K, Hassanian SM, Hashemy SI. PD-1/PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019;110:312-8.
  • 9
    Gupta R, Sinha S, Paul RN. The impact of microsatellite stability status in colorectal cancer. Curr. Probl. Cancer. 2018;42:548-59.
  • 10
    Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, et al. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann. Oncol. 2019;30:1096-03.
  • 11
    Miyauchi T, Yaguchi T, Kawakami Y. Inter-patient and intra-tumor heterogeneity in the sensitivity to tumor-targeted immunity in colorectal cancer. Nihon Rinsho. Meneki. Gakkai. Kaishi. 2017;40:54-9.
  • 12
    Cohen R, Pellat A, Boussion H, Svrcek M, Lopez-Trabada D, Trouilloud I, et al. Immunotherapy and metastatic colorectal cancers with microsatellite instability or mismatch repair deficiency. Bull Cancer. 2019;106:137-42.
  • 13
    Fabrizio DA, George TJJr, Dunne RF, Frampton G, Sun J, Gowen K, et al. Beyond microsatellite testing: assessment of tumor mutational burden identifies subsets of colorectal cancer who may respond to immune checkpoint inhibition. J. Gastrointest. Oncol. 2018;9:610-7.
  • 14
    Viale G, Trapani D, Curigliano G. Mismatch repair deficiency as a predictive biomarker for immunotherapy efficacy. Biomed. Res. Int. 2017;2017:4719194.
  • 15
    Lee V, Murphy A, Le DT, Diaz LA. Mismatch repair deficiency and response to immune checkpoint blockade. Oncologist. 2016;21:1200-11.
  • 16
    Zhang Y, Xu J, Zhang N, Chen M, Wang H, Zhu D. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett. 2019;458:123-35.
  • 17
    Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol. Res. 2019;7:1570-3.
  • 18
    Kong P, Wang J, Song Z, Liu S, He W, Jiang C, et al. Circulating lymphocytes, PD-L1 expression on tumor-infiltrating lymphocytes, and survival of colorectal cancer patients with different mismatch repair gene status. J. Cancer. 2019;10:1745-54.
  • 19
    Wu X, Gu Z, Chen Y, Chen B, Chen W, Weng L, et al. Application of PD-1 blockade in cancer immunotherapy. Comput. Struct. Biotechnol. J. 2019;17:661-74.
  • 20
    Kim JH, Park HE, Cho NY, Lee HS, Kang GH. Characterisation of PD-L1-positive subsets of microsatellite-unstable colorectal cancers. Br. J. Cancer. 2016;115:490-6.
  • 21
    Messersmith WA. NCCN Guidelines Updates: management of metastatic colorectal cancer. J. Natl. Compr. Canc. Netw. 2019;17:599-601.
  • 22
    Dawood S. The evolving role of immune oncology in colorectal cancer. Chin. Clin. Oncol. 2018;7:17.
  • 23
    Salem Me, Puccini A, Grothey A, Raghavan D, Goldberg Rm, Xiu J, et al. Landscape of tumor mutation load, mismatch repair deficiency, and PD-L1 expression in a large patient cohort of gastrointestinal cancers. Mol. Cancer Res. 2018;16:805-12.
  • 24
    Quiroga D, Lyerly HK, Morse MA. Deficient mismatch repair and the role of immunotherapy in metastatic colorectal cancer. Curr. Treat. Options Oncol. 2016;17:41.
  • 25
    Kather JN, Halama N, Jaeger D. Genomics and emerging biomarkers for immunotherapy of colorectal cancer. Semin. Cancer Biol. 2018;52:189-97.
  • 26
    Marginean EC, Melosky B. Is there a role for programmed death ligand-1 testing and immunotherapy in colorectal cancer with microsatellite instability? Part II-the challenge of programmed death ligand-1 testing and its role in microsatellite instability-high colorectal cancer. Arch. Pathol. Lab. Med. 2018;142:26-34.
  • 27
    Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat. Rev. Gastroenterol. Hepatol. 2019;16:361-75.
  • 28
    Lee KS, Kwak Y, Ahn S, Shin E, Oh HK, Kim DW, et al. Prognostic implication of CD274 (PD-L1) protein expression in tumor-infiltrating immune cells for microsatellite unstable and stable colorectal cancer. Cancer Immunol. Immunother. 2017;66:927-39.
  • 29
    Ho HL, Chou TY, Yang SH, Jiang JK, Chen WS, Chao Y, et al. PD-L1 is a double-edged sword in colorectal cancer: the prognostic value of PD-L1 depends on the cell type expressing PD-L1. J. Cancer Res. Clin. Oncol. 2019;145:1785-94.
  • 30
    Hamad T, Soong TR, Masugi Y, Kosumi K, Nowak JA, da Silva A, et al. TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PD-L1) expression status and tumor-infiltrating lymphocytes in colorectal carcinomas. Oncoimmunology. 2018;7:e1442999.
  • 31
    Maby P, Tougeron D, Hamieh M, Mlecnik B, Kora H, Bindea G, et al. Correlation between Density of CD8+ T-cell infiltrate in microsatellite unstable colorectal cancers and frameshift mutations: a rationale for personalized immunotherapy. Cancer Res. 2015;75:3446-55.
  • 32
    Riley JM, Cross AW, Paulos CM, Rubinstein MP, Wrangle J, Camp ER. The clinical implications of immunogenomics in colorectal cancer: A path for precision medicine. Cancer. 2018;124:1650-9.
  • 33
    Llosa NJ, Luber B, Siegel N, Awan AH, Oke T, Zhu Q, et al. Immunopathologic stratification of colorectal cancer for checkpoint blockade immunotherapy. Cancer Immunol. Res. 2019;7:1574-9.
  • 34
    Morse MA, Hochster H, Benson A. Perspectives on treatment of metastatic colorectal cancer with immune checkpoint inhibitor therapy. Oncologist. 2020;25:33-45.
  • 35
    Franke AJ, Skelton WP, Starr JS, Parekh H, Lee JJ, Overman MJ, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J. Natl. Cancer Inst. 2019;111:1131-41.
  • 36
    Arora SP, Mahalingam D. Immunotherapy in colorectal cancer: for the select few or all?. J. Gastrointest. Oncol. 2018;9:170-9.
  • 37
    Modest DP, Pant S, Sartore-Bianchi A. Treatment sequencing in metastatic colorectal cancer. Eur. J. Cancer. 2019;109:70-83.
  • 38
    Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, et al. 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;18:1182-91.
  • 39
    Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, et al. Durable clinical benefit with nivolumab plus ipilimumab in dna mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J. Clin. Oncol. 2018;36:773-9.
  • 40
    Ciardiello D, Vitiello PP, Cardone C, Martini G, Troiani T, Martinelli E, et al. Immunotherapy of colorectal cancer: Challenges for therapeutic efficacy. Cancer Treat. Rev. 2019;76:22-32.
  • 41
    O’Neil BH, Wallmark JM, Lorente D, Elez E, Raimbourg J, Gomez-Roca C, et al. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced colorectal carcinoma. PLoS One. 2017;12:e0189848.
  • 42
    Stein A, Folprecht G. Immunotherapy of colon cancer. Oncol. Res. Treat. 2018;41:282-5.
  • 43
    Yeon Yeon S, Jung SH, Jo YS, Choi EJ, Kim MS, Chung YJ, et al. Immune checkpoint blockade resistance-related B2M hotspot mutations in microsatellite-unstable colorectal carcinoma. Pathol. Res. Pract. 2019;215:209-14.
  • 44
    Tauber M, Cohen R, Laly P, Josselin L, Andre T, Mekinian A. Severe necrotizing myositis associated with long term anti-neoplastic efficacy following nivolumab plus ipilimumab combination therapy. Clin. Rheumatol. 2019;38:601-2.
  • 45
    Wilt C, Le DT. Integrating immunotherapy into colorectal cancer care. Oncology (Williston Park). 2018;32:494-8.
  • 46
    Chu JN, Choi J, Ostvar S, Torchia JA, Reynolds KL, Tramontano A, et al. Cost-effectiveness of immune checkpoint inhibitors for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Cancer. 2019;125:278-89.
  • 47
    Rawla P, Barsouk A, Hadjinicolaou AV, Barsouk A. Immunotherapies and targeted therapies in the treatment of metastatic colorectal cancer. Med. Sci. (Basel). 2019;7.

Publication Dates

  • Publication in this collection
    07 Dec 2020
  • Date of issue
    Oct-Dec 2020

History

  • Received
    9 Apr 2020
  • Accepted
    7 June 2020
  • Published
    13 Sept 2020
Sociedade Brasileira de Coloproctologia Av. Marechal Câmara, 160/916, 20020-080 Rio de Janeiro/RJ Brasil, Tel.: (55 21) 2240-8927, Fax: (55 21) 2220-5803 - Rio de Janeiro - RJ - Brazil
E-mail: sbcp@sbcp.org.br