Molecular classification and precision therapy of cancer: immune checkpoint inhibitors

Yingyan Yu

Front. Med. ›› 2018, Vol. 12 ›› Issue (2) : 229 -235.

PDF (155KB)
Front. Med. ›› 2018, Vol. 12 ›› Issue (2) : 229 -235. DOI: 10.1007/s11684-017-0581-0
COMMENTARY
COMMENTARY

Molecular classification and precision therapy of cancer: immune checkpoint inhibitors

Author information +
History +
PDF (155KB)

Abstract

On May 23, 2017, the US Food and Drug Administration (FDA) approved a treatment for cancer patients with positive microsatellite instability-high (MSI-H) markers or mismatch repair deficient (dMMR) markers. This approach is the first approved tumor treatment using a common biomarker rather than specified tumor locations in the body. FDA previously approved Keytruda for treatment of several types of malignancies, such as metastatic melanoma, metastatic non-small-cell lung cancer, recurrent or metastatic head and neck cancer, refractory Hodgkin lymphoma, and urothelial carcinoma, all of which carry positive programmed death-1/programmed death-ligand 1 biomarkers. Therefore, indications of Keytruda significantly expanded. Several types of malignancies are disclosed by MSI-H status due to dMMR and characterized by increased neoantigen load, which elicits intense host immune response in tumor microenvironment, including portions of colorectal and gastric carcinomas. Currently, biomarker-based patient selection remains a challenge. Pathologists play important roles in evaluating histology and biomarker results and establishing detection methods. Taking gastric cancer as an example, its molecular classification is built on genome abnormalities, but it lacks acceptable clinical characteristics. Pathologists are expected to act as “genetic interpreters” or “genetic translators” and build a link between molecular subtypes with tumor histological features. Subsequently, by using their findings, oncologists will carry out targeted therapy based on molecular classification.

Keywords

molecular classification / precision medicine / pembrolizumab / PD-1/PD-L1 / MSI-H

Cite this article

Download citation ▾
Yingyan Yu. Molecular classification and precision therapy of cancer: immune checkpoint inhibitors. Front. Med., 2018, 12(2): 229-235 DOI:10.1007/s11684-017-0581-0

登录浏览全文

4963

注册一个新账户 忘记密码

Insights into US FDA news

On May 23, 2017, the US Food and Drug Administration (FDA) approved treatment of cancer patients with positive microsatellite instability-high (MSI-H) markers or mismatch repair deficient (dMMR) markers (https://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm560167.htm). For the first time, the agency has approved a cancer treatment based on common biomarkers rather than tumor locations in the body. In September 2014, the FDA approved Keytruda for treatment of several types of malignancies, such as metastatic melanoma, metastatic non-small-cell lung cancer, recurrent or metastatic head and neck cancer, refractory classical Hodgkin lymphoma, and urothelial carcinoma. The recent approval remarkably expanded indications of Keytruda.

“This is an important first for the cancer community,” said Richard Pazdur, M.D., acting director of the Office of Hematology and Oncology Products in the FDA’s Center for Drug Evaluation and Research and director of FDA’s Oncology Center of Excellence. “Until now, the FDA has approved cancer treatments based on where in the body the cancer started—for example, lung or breast cancers. We have now approved a drug based on a tumor’s biomarker without regard to the tumor’s original location.” Keytruda functions through targeting the cellular pathway known as programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1), the proteins found on the body’s immune cells and some cancer cells, and blocking this pathway, thus helping the body’s immune system to fight cancer cells. Therefore, patients with positive PD-1/PD-L1 biomarkers and MSI-H or dMMR biomarkers will benefit from Keytruda treatment.

PD-1/PD-L1 pathway and MSI status for cancers

Next-generation sequencing provides an opportunity to characterize genome-wide variations with excellent resolution. Genomic aberrations can be specific to short DNA segments or span over many kb of DNA. These genomic events include mutations in protein-coding genes, which lead to either activation of oncogenes or inactivation of tumor suppressors [1]. High-throughput technologies can uncover genomic alterations that are shared across different tumor types, providing foundation for classification of cancer based on molecular aberrations [2]. Tumor classification based on recurrent genetic or epigenetic alterations that converge on unique pathways in different tumors can potentially provide another possibility for cancer treatment. In recent years, the cancer research community has exerted efforts on molecular classification of various types of cancers. Molecular classification determines gene mutations/variants using high-throughput technologies and stratifies tumors by gene expression profiles or gene mutations. Since the use of first-generation sequencing (Sanger sequencing) for mutation studies in 1975, considerable number of mutations have been discovered for different cancers (http://cancer.sanger.ac.uk/cosmic). Many potential targets have also been identified (http://archive.broadinstitute.org/cancer/cga/target). Binding of PD-L1 or PD-L2 to PD-1 can block T cell-mediated immune response to tumor cells. Antibodies that target PD-1 will disturb the ligand–receptor interface, activating T cells to attack tumors and intensify antitumor immune response (Fig. 1). PD-1 and PD-L1 (CD274) are targets for immunological checkpoints. Several types of cancers and immune cells express PD-L1, which plays a crucial role in inhibiting cancer immunity through binding with PD-1. Tumoral PD-L1 expression status has been demonstrated to be prognostic in multiple tumor types, including non-small-cell lung cancer, melanoma, renal cell carcinoma, nasopharyngeal carcinoma, and gastric cancer [3,4]. Monoclonal antibodies that disrupt PD-1/PD-L1 interaction have demonstrated favorable activity in different types of cancer as single and combinational therapies [510].

In several types of malignancies, MSI-H tumors due to dMMR are characterized by increased neoantigen load, which elicits intense host immune response in tumor microenvironment [1114]. Genomic features of tumors are linked to antitumor immunity status. MSI is a type of gene alteration characterized by nucleotide length abnormalities, which occur in tandem repeat units of 1 bp to 6 bp microsatellite. Given the deficiency or inactivation of one or more DNA MMR proteins (mutL homolog 1 (MLH1), mutS homolog 2 (MSH2), mutS homolog 6 (MSH6), or postmeiotic segregation increased 2 (PMS2)), cancer cells fail to repair DNA replication errors, leading to appearance of new alleles that are absent in normal DNA. Nowadays, examination of MSI status highly relies on histopathological observation, detection of a set of microsatellite markers for short nucleotide repeats (BAT25, BAT26, D2S123, D5S346, and D17S250 or a Promega MSI Analysis System), or immunohistochemistry for DNA MMR proteins, such as MLH1, MSH2, MSH6, and PMS2 [15]. In a Japanese study, MSI-H was detected in 17.7% of gastric cancer, and the highest proportions were observed in solid-type, poorly differentiated adenocarcinoma (43.0%) and papillary adenocarcinoma (32.5%) [16]. For large intestinal carcinoma, MMR rarely occurs in rectal cancers. MMR cancer accounts for 10%–15% of sporadic colon cancers. Large intestinal carcinoma with MSI phenotype is associated with proximal primary tumor location, high grade, mucinous pathology, and early stage. Most sporadic MSI tumors arise from sessile and serrated adenomas or polyps [17]. By contrast, dMMR has been reported in 20%–30% of endometrial cancers [18]. In addition, women with Lynch syndrome feature a lifetime risk of 8% for ovarian cancer [19,20]. For melanoma, MSI has been reported to be present in 2%–30% of primary tumors and 20%–77% of metastatic lesions [2123]. MSI-H exists in approximately 10%–20% of gastric cancer. Such tumors are associated with older patients, distal location, lower pathological tumor-node-metastasis stage, solid or intestinal subtype, and decreased lymph node metastasis. MSI-H gastric cancer also correlates well with prognosis [2426].

Assessing cancer cells that carry drug targets

Immune checkpoint inhibitors against PD-1/PD-L1 have disclosed remarkable therapeutic activity in solid tumors. However, biomarker-based patient selection remains a challenge. Expression of PD-l on immune cells is a promising biomarker. PD-l expression has been shown to predict remarkable benefits for anti-PD-1/PD-L1 agents. However, absence of PD-1 does not preclude treatment efficacy [27]. PD-L1 expression in tumor-associated immune cells has also been shown to play a role in predicting treatment response in some studies. PD-L1 expression in pre-treatment tumor microenvironment enriches response to anti-PD-1/PD-L1 therapies. Sunshine et al. (2016) reported that immunohistochemical staining characteristics of antibody 5H1, SP142, 28-8, SP263, and 22C3 clones used in PD-L1 assays were very similar to those of melanoma specimens when other assay conditions were held constant [28]. Among these antibodies, the 22C3 clone is an FDA-approved reagent for selecting non-small-cell lung cancer patients for pembrolizumab treatment [29]. Pathologists play a relevant role with regard to patient selection. Garon et al. (2016) proposed that in non-small-cell lung cancer, patients whose tumors feature over 50% malignant cells showing PD-L1 expression (using 22C3 antibody) will more likely respond to pembrolizumab than those with less than 50% malignant cells [30].

In examination of DNA mismatch repair status, many groups recommended usage of at least four different molecular markers in combination with immunohistochemistry. MSI in gastrointestinal cancer is commonly associated with loss of MLH1/PMS2 expression. In the MMR system, four genes, including MLH1, MSH2, MSH6, and PMS2, play important roles; these genes were named as such because of their homology to E. coli MMR genes. Immunohistochemistry is widely used for MMR examination. Tumors with dMMR will usually show complete loss of expression of one or more wild-type proteins. Detecting all four proteins provides further information in determining defective proteins. In general, MLH1 and PMS2 form a heterodimer complex in cells. Loss of PMS2 expression alone indicates a PMS2 gene defect. However, combined losses of PMS2 and MLH1 suggest that the defect lies in MLH1, which is responsible for stability of PMS2. Similarly, loss of MSH6 represents an MSH6 defect alone, whereas losses of MSH6 and MSH2 protein expressions indicate a defect in MSH2 gene [3133].

For genetic assay, tumors were designated as MSI-H when at least two out of the five markers (BAT25, BAT26, D2S123, D5S346, D17S250, or a Promega MSI Analysis System) showed instability and as MSS when none showed instability. MSI-low cases showed instability in only one marker. Promega MSI Analysis System is a commercially available kit consisting of five mononucleotide markers (BAT25, BAT26, MONO-27, NR-21, and NR-24) with high sensitivity and specificity for detecting MSI-H phenotypes [34,35]. Although polymerase chain reaction-based molecular examination has been regarded the standard diagnostic method in assessing MSI status in many tumors, immunohistochemistry is more simple, rapid, and universally available than molecular testing.

Keytruda expanded indications for gastric cancer therapy

In September 2014, a four-category molecular classification of gastric cancer was proposed by The Cancer Genome Atlas (TCGA) research network, which reported a panorama of molecular profiles for 295 cases of gastric cancer. Merits of the TCGA study included integrated data of 295 cases of gastric cancer from array-based copy number analysis, whole-genome sequencing, array-based methylation profiling, mRNA sequencing, microRNA sequencing, and reverse-phase protein array. The TCGA group divided gastric cancer into Epstein-Barr virus (EBV)-positive subtype, MSI subtype, genomically stable (GS) subtype, and chromosomal instability subtype (CIN). Among these types, carcinoma of EBV-positive subtype showed significant mutated event of PIK3CA and a novel recurrent amplification at 9p24.1, resulting in overexpression of PD-L1 and PD-L2. To confirm EBV infection, detection of EBV-encoded RNA probe can be used. Carcinoma of MSI subtype revealed high frequency of gene mutation. Carcinoma of GS subtype was mainly observed in diffused-type histology (poorly cohesive) with RHOA gene mutation or a fusion gene of CLDN18-ARHGAP26, which contributes to invasive biological behavior of carcinoma cells. CIN subtype carcinoma is characterized by aneuploidy and focal gene amplification of receptor tyrosine kinase [36]. In TCGA classification, EBV-positive (9%) and MSI subtypes (22%) of gastric cancer serve as indicators for Keytruda targeted drug, indicating that at least 31% of gastric cancer cases will benefit from Keytruda treatment. EBV-positive subtype showed extensive immune cell infiltration and PD-L1/PD-L2 overexpression. Therefore, PD-L1/PD-L2 antagonists may be effective, whereas for MSI subtype, Keytruda treatment is recommended.

TCGA molecular classification is built on genome abnormalities and lacks acceptable clinical characteristics. Difficulty arises from using this approach in routine clinical practice. To solve this problem, pathologists are expected to act as “genetic interpreters” or “genetic translators.” They can build a link between molecular subtypes using their histological features and describe them on pathological reports. Subsequently, by using their findings, oncologists will carry out targeted therapy based on molecular classification. Undoubtedly, pathologists will play important roles in promoting clinical translation of new molecular classifications. For this purpose, the author reviewed glass slides of 295 cases of gastric cancer uploaded in public cancer databases (http://www.cbioportal.org/study?ID=stad_tcga_pub#clinical). Among these samples, glass slides of 227 cases (76.95%) came from paraffin-embedded tissues, and the remaining 68 cases (23.05%) were from frozen sections. In the TCGA set, a majority of the EBV-positive subtype shows lymphoepithelial lesions on the background of moderately or poorly differentiated tubular adenocarcinomas. MSI subtype shows expansive medullary or solid pattern with a pushing forward histology in invasive frontier. The invasive frontier is surrounded by numerous lymphocytes. A minority of papillary carcinomas, tubular adenocarcinomas, or miscellaneous carcinomas are also categorized under the MSI subtype. GS subtype dominantly corresponds to poorly cohesive carcinomas, including signet-ring cell carcinoma and its variants, such as small-cell signet-ring cell carcinoma, large-cell signet-ring cell carcinoma, and mucinous signet-ring cell carcinoma. CIN subtype includes half of the cases in the TCGA group and is more complex in morphologic phenotypes (Table 1). Preferably, pathologists must use pathological language to interpret the TCGA molecular classification on their clinical reports. As EBV-positive or MSI subtype presents a relatively characteristic histology, an experienced pathologist can offer confirmative diagnosis for these subtypes of gastric cancer based on histological observations.

Possible mechanisms for Keytruda resistance

Monoclonal antibodies that inhibit the PD-1/PD-L1 pathway are clinically active against a broad range of tumors; however, some tumor cells respond less because of innate resistance (innate anti-PD-1 resistance signature) and the presence of somatic differences in tumor cells (constitutive activation of β-catenin or loss of phosphatase and tensin homolog) that inhibit activation and recruitment of T cells to the tumor microenvironment [3740]. These mechanisms will result in treatment failure for patients who do not respond to current checkpoint antibodies; moreover, identifying drug-resistant patients is beyond the limited ability of the current FDA-approved PD-L1 companion immunohistochemical assays [29]. For instance, in a melanoma patient who responds well to anti-PD-1 therapy, delayed relapses will occur long after initial objective tumor regression despite continuous therapy. Recently, Zaretsky and coworkers analyzed biopsy samples from paired baseline and relapsing lesions in four patients with metastatic melanoma. The patients initially responded to anti-PD-1 therapy (pembrolizumab) but experienced disease progression months to years later. By using whole-exome sequencing, two patients were diagnosed with loss-of-function mutations in interferon-receptor-associated JAK1 or JAK2, resulting in drug resistance. Truncating mutation in β-2-microglobulin (B2M) was identified in the third patient. These gene mutations resulted in insensitivity to antiproliferative effects on cancer cells and lack of response to interferon γ. Truncating mutation of B2M led to loss of surface expression of major histocompatibility complex class I. Therefore, acquired resistance to PD-1 blockade immunotherapy in patients with melanoma is associated with defects in pathways involved in interferon-receptor signaling and antigen presentation [41,42]. Although the new targeted drug features wide indications, existence of therapeutic resistance should be considered. Researchers should develop methods for detecting resistant patients and exploring new therapeutic targets for them.

References

[1]

Chakravarthi BVSK, Nepal S, Varambally S. Genomic and epigenomic alterations in cancer. Am J Pathol 2016; 186(7): 1724–1735

[2]

Hoadley KA, Yau C, Wolf DM, Cherniack AD, Tamborero D, Ng S, Leiserson MD, Niu B, McLellan MD, Uzunangelov V, Zhang J, Kandoth C, Akbani R, Shen H, Omberg L, Chu A, Margolin AA, Van’t Veer LJ, Lopez-Bigas N, Laird PW, Raphael BJ, Ding L, Robertson AG, Byers LA, Mills GB, Weinstein JN, Van Waes C, Chen Z, Collisson EA; Cancer Genome Atlas Research Network, Benz CC, Perou CM, Stuart JM. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 2014; 158(4): 929–944

[3]

Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, Kohrt HE, Horn L, Lawrence DP, Rost S, Leabman M, Xiao Y, Mokatrin A, Koeppen H, Hegde PS, Mellman I, Chen DS, Hodi FS. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014; 515(7528): 563–567

[4]

Zhang J, Fang W, Qin T, Yang Y, Hong S, Liang W, Ma Y, Zhao H, Huang Y, Xue C, Huang P, Hu Z, Zhao Y, Zhang L. Co-expression of PD-1 and PD-L1 predicts poor outcome in nasopharyngeal carcinoma. Med Oncol 2015; 32(3): 86

[5]

Twyman-Saint Victor C, Rech AJ, Maity A, Rengan R, Pauken KE, Stelekati E, Benci JL, Xu B, Dada H, Odorizzi PM, Herati RS, Mansfield KD, Patsch D, Amaravadi RK, Schuchter LM, Ishwaran H, Mick R, Pryma DA, Xu X, Feldman MD, Gangadhar TC, Hahn SM, Wherry EJ, Vonderheide RH, Minn AJ. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015; 520(7547): 373–377

[6]

Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, Martins R, Eaton K, Chen S, Salay TM, Alaparthy S, Grosso JF, Korman AJ, Parker SM, Agrawal S, Goldberg SM, Pardoll DM, Gupta A, Wigginton JM. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366(26): 2455–2465

[7]

Kim JW, Eder JP. Prospects for targeting PD-1 and PD-L1 in various tumor types. Oncology (Williston Park) 2014; 28(Suppl 3): 15–28

[8]

Weinstock M, McDermott D. Targeting PD-1/PD-L1 in the treatment of metastatic renal cell carcinoma. Ther Adv Urol 2015; 7(6): 365–377

[9]

Taube JM, Klein A, Brahmer JR, Xu H, Pan X, Kim JH, Chen L, Pardoll DM, Topalian SL, Anders RA. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 2014; 20(19): 5064–5074

[10]

Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol 2015; 33(17): 1974–1982

[11]

Li SK, Martin A. Mismatch repair and colon cancer: mechanisms and therapies explored. Trends Mol Med 2016; 22(4): 274–289

[12]

Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, Blosser RL, Fan H, Wang H, Luber BS, Zhang M, Papadopoulos N, Kinzler KW, Vogelstein B, Sears CL, Anders RA, Pardoll DM, Housseau F. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov 2015; 5(1): 43–51

[13]

Dudley JC, Lin MT, Le DT, Eshleman JR. Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res 2016; 22(4): 813–820

[14]

Yoon K, Lee S, Han TS, Moon SY, Yun SM, Kong SH, Jho S, Choe J, Yu J, Lee HJ, Park JH, Kim HM, Lee SY, Park J, Kim WH, Bhak J, Yang HK, Kim SJ. Comprehensive genome- and transcriptome-wide analyses of mutations associated with microsatellite instability in Korean gastric cancers. Genome Res 2013; 23(7): 1109–1117

[15]

Richman S. Deficient mismatch repair: read all about it. Int J Oncol 2015; 47(4): 1189–1202

[16]

Arai T, Sakurai U, Sawabe M, Honma N, Aida J, Ushio Y, Kanazawa N, Kuroiwa K, Takubo K. Frequent microsatellite instability in papillary and solid-type, poorly differentiated adenocarcinomas of the stomach. Gastric Cancer 2013; 16(4): 505–512

[17]

Sinicrope FA, Rego RL, Foster N, Sargent DJ, Windschitl HE, Burgart LJ, Witzig TE, Thibodeau SN. Microsatellite instability accounts for tumor site-related differences in clinicopathologic variables and prognosis in human colon cancers. Am J Gastroenterol 2006; 101(12): 2818–2825

[18]

Matthews KS, Estes JM, Conner MG, Manne U, Whitworth JM, Huh WK, Alvarez RD, Straughn JM Jr, Barnes MN, Rocconi RP. Lynch syndrome in women less than 50 years of age with endometrial cancer. Obstet Gynecol 2008; 111(5): 1161–1166

[19]

Engel C, Loeffler M, Steinke V, Rahner N, Holinski-Feder E, Dietmaier W, Schackert HK, Goergens H, von Knebel Doeberitz M, Goecke TO, Schmiegel W, Buettner R, Moeslein G, Letteboer TG, Gómez García E, Hes FJ, Hoogerbrugge N, Menko FH, van Os TA, Sijmons RH, Wagner A, Kluijt I, Propping P, Vasen HF. Risks of less common cancers in proven mutation carriers with lynch syndrome. J Clin Oncol 2012; 30(35): 4409–4415

[20]

Watson P, Vasen HF, Mecklin JP, Bernstein I, Aarnio M, Järvinen HJ, Myrhøj T, Sunde L, Wijnen JT, Lynch HT. The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int J Cancer 2008; 123(2): 444–449

[21]

Talwalkar VR, Scheiner M, Hedges LK, Butler MG, Schwartz HS. Microsatellite instability in malignant melanoma. Cancer Genet Cytogenet 1998; 104(2): 111–114

[22]

Alvino E, Marra G, Pagani E, Falcinelli S, Pepponi R, Perrera C, Haider R, Castiglia D, Ferranti G, Bonmassar E, Jiricny J, Zambruno G, D’Atri S. High-frequency microsatellite instability is associated with defective DNA mismatch repair in human melanoma. J Invest Dermatol 2002; 118(1): 79–86

[23]

Palmieri G, Ascierto PA, Cossu A, Colombino M, Casula M, Botti G, Lissia A, Tanda F, Castello G. Assessment of genetic instability in melanocytic skin lesions through microsatellite analysis of benign naevi, dysplastic naevi, and primary melanomas and their metastases. Melanoma Res 2003; 13(2): 167–170

[24]

Beghelli S, de Manzoni G, Barbi S, Tomezzoli A, Roviello F, Di Gregorio C, Vindigni C, Bortesi L, Parisi A, Saragoni L, Scarpa A, Moore PS. Microsatellite instability in gastric cancer is associated with better prognosis in only stage II cancers. Surgery 2006; 139(3): 347–356

[25]

Fang WL, Chang SC, Lan YT, Huang KH, Chen JH, Lo SS, Hsieh MC, Li AF, Wu CW, Chiou SH. Microsatellite instability is associated with a better prognosis for gastric cancer patients after curative surgery. World J Surg 2012; 36(9): 2131–2138

[26]

Lee HS, Choi SI, Lee HK, Kim HS, Yang HK, Kang GH, Kim YI, Lee BL, Kim WH. Distinct clinical features and outcomes of gastric cancers with microsatellite instability. Mod Pathol 2002; 15(6): 632–640

[27]

Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G Jr, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled-Filhart M, Garon EB. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016; 387(10027): 1540–1550

[28]

Sunshine JC, Nguyen PL, Kaunitz GJ, Cottrell TR, Berry S, Esandrio J, Xu H, Ogurtsova A, Bleich KB, Cornish TC, Lipson EJ, Anders RA, Taube JM. PD-L1 expression in melanoma: a quantitative immunohistochemical antibody comparison. Clin Cancer Res 2017; 23(16):4938–4944

[29]

Ma W, Gilligan BM, Yuan J, Li T. Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy. J Hematol Oncol 2016; 9(1): 47

[30]

Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Patnaik A, Aggarwal C, Gubens M, Horn L, Carcereny E, Ahn MJ, Felip E, Lee JS, Hellmann MD, Hamid O, Goldman JW, Soria JC, Dolled-Filhart M, Rutledge RZ, Zhang J, Lunceford JK, Rangwala R, Lubiniecki GM, Roach C, Emancipator K, Gandhi L; KEYNOTE-001 Investigators. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 2015; 372(21): 2018–2028

[31]

Mensenkamp AR, Vogelaar IP, van Zelst-Stams WA, Goossens M, Ouchene H, Hendriks-Cornelissen SJ, Kwint MP, Hoogerbrugge N, Nagtegaal ID, Ligtenberg MJ. Somatic mutations in MLH1 and MSH2 are a frequent cause of mismatch-repair deficiency in Lynch syndrome-like tumors. Gastroenterology 2014; 146(3): 643–646.e8

[32]

Fadhil W, Ilyas M. Immunostaining for mismatch repair (MMR) protein expression in colorectal cancer is better and easier to interpret when performed on diagnostic biopsies. Histopathology 2012; 60(4): 653–655

[33]

Bao F, Panarelli NC, Rennert H, Sherr DL, Yantiss RK. Neoadjuvant therapy induces loss of MSH6 expression in colorectal carcinoma. Am J Surg Pathol 2010; 34(12): 1798–1804

[34]

Xicola RM, Llor X, Pons E, Castells A, Alenda C, Piñol V, Andreu M, Castellví-Bel S, Payá A, Jover R, Bessa X, Girós A, Duque JM, Nicolás-Pérez D, Garcia AM, Rigau J, Gassull MA; Gastrointestinal Oncology Group of the Spanish Gastroenterological Association. Performance of different microsatellite marker panels for detection of mismatch repair-deficient colorectal tumors. J Natl Cancer Inst 2007; 99(3): 244–252

[35]

Bapat B, Lindor NM, Baron J, Siegmund K, Li L, Zheng Y, Haile R, Gallinger S, Jass JR, Young JP, Cotterchio M, Jenkins M, Grove J, Casey G, Thibodeau SN, Bishop DT, Hopper JL, Ahnen D, Newcomb PA, Le Marchand L, Potter JD, Seminara D; Colon Cancer Family Registry. The association of tumor microsatellite instability phenotype with family history of colorectal cancer. Cancer Epidemiol Biomarkers Prev 2009; 18(3): 967–975

[36]

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014; 513(7517): 202–209

[37]

Bifulco CB, Urba WJ. Unmasking PD-1 resistance by next-generation sequencing. N Engl J Med 2016; 375(9): 888–889

[38]

Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, Berent-Maoz B, Pang J, Chmielowski B, Cherry G, Seja E, Lomeli S, Kong X, Kelley MC, Sosman JA, Johnson DB, Ribas A, Lo RS. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016; 165(1): 35–44

[39]

Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic b-catenin signalling prevents anti-tumour immunity. Nature 2015; 523(7559): 231–235

[40]

Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, Xu C, McKenzie JA, Zhang C, Liang X, Williams LJ, Deng W, Chen G, Mbofung R, Lazar AJ, Torres-Cabala CA, Cooper ZA, Chen PL, Tieu TN, Spranger S, Yu X, Bernatchez C, Forget MA, Haymaker C, Amaria R, McQuade JL, Glitza IC, Cascone T, Li HS, Kwong LN, Heffernan TP, Hu J, Bassett RL Jr, Bosenberg MW, Woodman SE, Overwijk WW, Lizée G, Roszik J, Gajewski TF, Wargo JA, Gershenwald JE, Radvanyi L, Davies MA, Hwu P. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov 2016; 6(2): 202–216

[41]

Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, Torrejon DY, Abril-Rodriguez G, Sandoval S, Barthly L, Saco J, Homet Moreno B, Mezzadra R, Chmielowski B, Ruchalski K, Shintaku IP, Sanchez PJ, Puig-Saus C, Cherry G, Seja E, Kong X, Pang J, Berent-Maoz B, Comin-Anduix B, Graeber TG, Tumeh PC, Schumacher TN, Lo RS, Ribas A. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016; 375(9): 819–829

[42]

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(6349): 409–413

RIGHTS & PERMISSIONS

Higher Education Press and Springer-Verlag GmbH Germany

AI Summary AI Mindmap
PDF (155KB)

2914

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/