Aan dit artikel is enkele uren gewerkt. Beoordelen, opzoeken relevante informatie, uitleggen, vertalen en plaatsen op de website. Mocht u ons willen ondersteunen om kanker-actueel online te houden zodat we meer van dit soort artikelen kunnen bijven publiceren dan kunt u ons helpen via donaties: https://kanker-actueel.nl/NL/donaties.html of doneer al of niet anoniem op rekeningnummer NL79 RABO 0372931138 t.n.v. Stichting Gezondheid Actueel Terneuzen.

28 september 2017: Bron: J Cancer. 2016; 7(12): 1599–1604 en Biomed Res Int. 2017; 2017: 4346576.

Al eind jaren negentig en begin jaren 2000 werden ook in Nederland en België studies uitgevoerd met immuuntherapie bij darmkanker en andere vormen van spijsverteringskanker als alvleesklierkanker, en maagkanker en slokdarmkanker. Sommige met succes andere zonder succes. (Tekst gaat verder onder foto)

immuuntherapie
De belangrijkste studie naar de effecten van immuuntherpie was in 1999 in The Lancet gepubliceerde fase III studie van Vermorken J. B., Claessen A. M. E., Van Tinteren H., et al. Active specific immunotherapy for stage II and stage III human colon cancer: A randomised trial. Lancet. 1999;353(9150):345–350. doi: 10.1016/S0140-6736(98)07186-4 die ook jarenlang werd vermeld in oncoline maar daaruit inmiddels is verwijderd.

Maar in deze studie werd een 44% vermindering van het risico op een recidief gezien wanneer patiënten met darmkanker  werden behandeld met Oncovax (p = 0.023). In een subgroep van patienten met een stadium II van darmkanker was dat risico zelfs met 61% verminderd. Maar om welke reden dan ook is die aanpak vanuit die studie nooit echt doorgezet, zie onder autovaccinatie op onze site. Onterecht m.i.

Uit het studieverslag over Oncovax:

De werkzaamheid van OncoVAX werd vervolgens geëvalueerd in de adjuvante setting in drie fase III klinische studies waarbij patiënten gerandomiseerd werden ingedeeld om alleen een operatie te ondergaan  of een operatie gevolgd door een autovaccinatie met Oncovax.
De eerste studie (8102) werd in 1981 gestart en 98 patiënten met darmkanker in stadium II en III namen deel. De primaire einddoelen, overall overleving (OS) en ziektevrije overleving (DFS) werden niet bereikt (HR voor OS = 1,75, p = 0,68; HR voor DFS = 1,58, p = 0,147).
In de subgroepanalyses werd echter een statistisch significant voordeel van OncoVAX gezien bij patiënten met dikke darmkanker (HR voor OS = 2,83, p = 0,02; HR voor DFS = 2,67, p = 0,039) maar niet bij patiënten met rectale kanker (HR voor OS = 1,13, p = 0,772; HR voor DFS = 1,05, p = 0,905).

In de fase III 5283 studie met totaal 412 darmkankerpatiënten met stadium II en III werden er statistisch geen verschillen in overall overleving (OS) en ziektevrije tijd (DFS) waargenomen.

Tenslotte werden in de fase 8701 III studie 254 patiënten met darmkanker stadium II en III gerandomiseerd ingedeeld en onderzocht wat de resultaten op overall overleving (OS) en ziektevrije tijd (DFS) zouden zijn als het vaccin 4 keer werd toegediend in plaats van 3 keer. In die studie werd een 44% verminderd risico gezien op een recidief bij patiënten die werden behandeld met OncoVAX (p = 0,023). In de subgroepanalyses werd een nog betere werkzaamheid waargenomen bij patiënten met stadium II (61% risicoreductie op een recidief).

Een meta-analyse, inclusief de 3 hierboven vermelde onderzoeken, vertoonde een verbetering van het risico op een recidief door OncoVAX met een jaarlijkse kansverlaging van 25 ± 13% (p = 0,05). De subgroepanalyse per stadium vertoonde een statistisch significante verbetering bij stadium II patiënten (p = 0,05).

Een andere studie liet zien dat dendritische celtherapie met het Newcastle Disease Virus ook goede resultaten liet zien. Niet bij rectale vormen van kanker maar wel bij dikke darmkanker:

De resultaten van een gerandomiseerde fase III trial met een NDV autoloog gemodificeerd vaccin bij patiënten die een radicale resectie van levermetastasen vanuit darmkanker hadden ondergaan, werden eind 2000 gepubliceerd. Aan deze studie namen 51 patiënten deel. Er werden statistisch geen significante verschillen in overall overleving (OS) en in ziektevrije tijd (DFS) geconstateerd. Maar in de subgroepanalyses werd wel een statistisch significant voordeel waargenomen bij patiënten met dikke darmkanker met betrekking tot overall overleving (OS) (HR 3.3, p = 0.042) en ziektevrije tijd (DFS) (HR 2.7, p = 0.047), Maar ook hier niet bij patiënten met rectale kanker. Waarom niet wordt niet beschreven. Zie deze studie: 

Schulze T., Kemmner W., Weitz J., Wernecke K.-D., Schirrmacher V., Schlag P. M. Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: Results of a prospective randomized trial. Cancer Immunology, Immunotherapy. 2009;58(1):61–69

Inmiddels zijn naast de immuuntherapeutische studies met autovaccinatie en dendritische celtherapie en andere vaccins er ook andere aanpak met immuuntherapie met anti-PD medicijnen bijgekomen.

En met succes.

Zo blijkt uit de Checkmate 142 studie waarin o.a. nivolumab wel en niet wordt gecombineerd met ipilimumab:

72 patiënten behandeld met nivolumab gaven hoopgevende resultaten op 12 maanden meting met een ziektevrije overleving van 48,4% en overall overleving 73.8%. Zelfs 2 patienten bereikten binnen een jaar een complete remissie. 

Results: 70 (N3) and 30 (N3 + I1) MSI-H pts and 3 (N1 + I1), 10 (N1 + I3), and 10 (N3 + I1) non-MSI-H pts were enrolled. All non-MSI-H pts and 87% (N3) and 93% (N3 + I1) of MSI-H pts had ≥2 prior regimens. 47 (67%; N3) and 18 (60%; N3 + I1) MSI-H pts remain on tx. Efficacy data for MSI-H pts are shown in the Table. Responses were also seen in non-MSI-H pts. Median (95% CI) PFS across all non-MSI-H pts was 1.4 mo (1.2, 1.9). Responses were observed regardless of tumor PD-L1 expression. Treatment-related adverse events (TRAEs) occurred in 41 (59%; N3) and 25 (83%; N3 + I1) MSI-H pts; 10 (14%; N3) and 8 (27%; N3 + I1) pts had Grade 3–4 TRAEs. One pt on N3 had a Grade 5 TRAE (sudden death). Additional biomarker data including MSI assessment and influence of BRAF/KRAS mutations will be presented.

En immuuntherapie met een gemoduleerd virus liet op ESMO 2017 ook uitstekende resultaten zien, zoals we al eerder hebben gepubliceerd op kanker-actueel: 

https://kanker-actueel.nl/NL/immuuntherapie-met-het-gemoduleerde-virus-ankara5t4-trovax-plus-lage-dosis-cyclophosphamide-zorgt-voor-verdubbeling-van-mediane-overall-overleving-112-vs-20-maanden-bij-vergevorderde-darmkanker.html

Saemnvattend zien we dat immuuntherapie bij vormen van spijsverteringskanker op de goede weg is. Ook in Nederland en België.

Hier een aantal studies met immuuntherapeutische aanpak die al klaar zijn of die nog lopen. Klik op de NCT nummers voor studieprotocollen:

Table 1

Ongoing studies on gastric, gastroesophageal junction, and esophageal cancers.

NCT identifierSettingPhaseStudy interventionsNumber of patientsPrimary endpoint
Checkpoint inhibitors
NCT02689284 Metastatic HER2+ GC/GEJC Ib/II Margetuximab+ pembrolizumab 52 MTD and MAD for margetuximab; duration of response; 12-month ORR
NCT02563548 Metastatic GC after 1st line Ib PEGPH20 +pembrolizumab 81 DLT; 18-month ORR
NCT02443324 Metastatic GC/GEJC and other tumours I Ramucirumab + pembrolizumab 155 DLT
NCT02589496 Metastatic GC/GEJC after first line II Pembrolizumab 40 2-year RR
NCT02901301 First-line HER2 + GC Ib/II Pembrolizumab + trastuzumab + capecitabine + cisplatin 49 RP2D; 6-week ORR
NCT02954536 First-line HER2+ GC/GEJC/EC II Pembrolizumab + trastuzumab + capecitabine + cisplatin 37 6-month PFS
NCT02318901 Unresectable HER2 + GC/GEJC II Pembrolizumab + ado-trastuzumab emtansine 90 RP2D
NCT02559687 EC (adenocarcinoma or squamous cell)/GEJC after 2nd line II Pembrolizumab 100 2-year ORR
NCT02494583 First-line GC/GEJC III (random) Pembrolizumab versus pembrolizumab + cisplatin + 5-fluorouracil or capecitabine versus placebo + cisplatin + 5-FU or capecitabine 750 44-month PFS and OS
NCT02370498 Second-line GC/GEJC III (random) Pembrolizumab versus paclitaxel 720 PFS, OS
NCT02564263 EC (adenocarcinoma or squamous cell) /GEJC after 1st line III (random) Pembrolizumab versus investigator's choice of standard therapy (paclitaxel, docetaxel, or irinotecan) 600 3-year PFS and OS
NCT02872116 Unresectable GC/GEJC III (random) Nivolumab + ipilimumab versus nivolumab + oxaliplatin + fluoropyrimidine versus oxaliplatin + fluoropyrimidine 1266 40-month OS in patients PD-L1 +
NCT02864381 Metastatic GC/GEJC II (random) GS-5745 + nivolumab versus nivolumab alone 120 2-year ORR
NCT02340975 Pretreated metastatic/GC/GEJC Ib/II (random) MEDI4736 + tremelimumab versus MEDI4736 versus tremelimumab 135 Phase Ib: DTL,
Phase II: ORR and 6-month PFS
NCT02625623 3rd-line GC/GEJC III (random) Avelumab+ BSC versus chemotherapy (paclitaxel or irinotecan)+BSC or BSC alone 330 2-year OS
NCT02625610 1st-line GC/GEJC III (random) Maintenance with avelumab versus continuation of 1st-line chemotherapy 666 3-year OS and PFS

Immunotherapy + radiotherapy
NCT02642809 1st-line EC I Pembrolizumab + brachytherapy 15 Tolerability and toxicity
NCT02830594 Pretreated EC/GC(GEJC II Pembrolizumab + external beam palliative radiation therapy 14 Biomarkers
NCT02735239 Metastatic EC I/II Durvalumab + oxaliplatin/capecitabine 75 AE, DLT, laboratory evaluations
Vaccines
NCT02276300 Metastatic HER 2 + GC I HER2-derived peptide vaccination 12 Safety and tolerability
NCT02317471 Stage III gastric cancer I/II Vaccination with autologous tumour derived heat shock protein gp96 45 DFS
NCT02795988 Metastatic HER 2 + GC/GEJC Ib/II IMU-131 HER2/Neu peptide vaccine+ cisplatin and either 5-FU or capecitabine chemotherapy 18 RP2D, AE
Cytokines
NCT01691664 Locally advanced EC NS (random) Radiation therapy alone or with DC-CIK cellular therapy 40 DFS
NCT01691625 Locally advanced EC NS (random) Concurrent chemoradiation with or without DC-CIK 50 Quality of life
NCT02504229 Metastatic refractory GC II
(random)
Chemotherapy with or without DC-CIK 80 PFS
NCT01783951 Metastatic refractory GC I/II S-1 with or without DC-CIK 30 PFS

CAR-T cells
NCT02713984 Metastatic refractory HER 2 + GC I/II Anti-HER2 CAR-T cells 60 Toxicity
NCT02725125 Metastatic refractory GC I/II EPCAM-targeted CAR-T cells 19 DCR
NCT02617134 Metastatic refractory MUC1+ GC I/II Anti-MUC1 CAR-T cells 20 Toxicity
NCT02349724 Metastatic refractory CEA+ GC I Anti-CEACAR-T cells 75 Toxicity
NCT02862028 Metastatic refractory EGFR+ GC I/II Anti-PD-1CAR-T cells 20 ORR, DCR, OS, PFS
NCT03013712 Metastatic refractory EpCAM+ GC/EC I/II Anti-EpCAMCAR-T cells 60 Toxicity

GC, gastric cancer; GEJC, gastroesophageal junction cancer; EC: esophageal cancer; NA, not assessed; MTD, maximum tolerated dose; MAD, maximum administered dose; DTL, dose limiting toxicity; ORR, overall response rate; OS, overall survival; PFS, progression-free survival; DFS, disease-free survival; NS, not specified; RP2D, recommended dose of phase II; AE, adverse events; DCR, disease control rate; BSC, best supportive care; DCR disease control rate.

Het volledige studierapport: Current Status and Perspective of Immunotherapy in Gastrointestinal Cancers is gratis in te zien. Evenals het studierapport: Immunotherapy in Gastrointestinal Cancers

Abstracten van beide studies plus referentielijsten volgen onderaan dit artikel.

Lees ook dit artikel: 

https://kanker-actueel.nl/NL/pole-mutatie-veel-kankerpatienten-met-erfelijke-vormen-van-kanker-hebben-naast-een-p1-ligand-een-pole-mutatie-en-reageren-goed-op-immuuntherapie-met-anti-pd-medicijnen-checkpointremmers-als-pembrolizumab-en-nivolumab.html

En lees ook de richtlijnen voor het behandelen van uitgezaaide darmkanker  zoals gepubliceerd door de ESMO: 

Metastatic colorectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up

Interessant is ook deze review over zogeheten oncolytische virussen: 

Oncolytic viruses: From bench to bedside with a focus on safety

Lees ook dit artikel: 

The emerging role of immunotherapy in colorectal cancer

From a clinician perspective, the use of immunotherapies in recent clinical trials gave us the opportunity to contribute to a paradigmatic shift in the treatment of GI cancers. We are glad to observe highly pretreated patients experiencing a dramatic clinical benefit after treatment start, with symptoms relief, long lasting disease stabilization, and an overall manageable safety profile.

Biomed Res Int. 2017; 2017: 4346576.
Published online 2017 Jul 3. doi:  10.1155/2017/4346576
PMCID: PMC5512095

Immunotherapy in Gastrointestinal Cancers

Abstract

Gastrointestinal cancers represent a major public health problem worldwide. Immunotherapeutic strategies are currently under investigation in this setting and preliminary results of ongoing trials adopting checkpoint inhibitors are striking. Indeed, although a poor immunogenicity for GI has been reported, a strong biological rationale supports the development of immunotherapy in this field. The clinical and translational research on immunotherapy for the treatment of GI cancers started firstly with the identification of immune-related mechanisms possibly relevant to GI tumours and secondly with the development of immunotherapy-based agents in clinical trials. In the present review a general overview is firstly provided followed by a focus on major findings on gastric, colorectal, and hepatocellular carcinomas. Finally, pathological and molecular perspectives are provided since many efforts are ongoing in order to identify possible predictive biomarkers and to improve patients' selection. Many issues are still unsolved in this field; however, we strongly believe that immunotherapy might positively affect the natural history of a subgroup of GI cancer patients improving outcome and the overall quality of life.

Finally the most efforts are focusing on the development of novel approaches to enhance this innovative strategy. All ongoing trials are shown in Tables Tables1113. Promising trials have been evaluating innovative combination treatments (so-called “combo-immunotherapy”), that is, PD-1 or PD-L1 blockade in combination with (1) anti-CTLA4, (2) adaptive immunotherapy such as anti-LAG3, (3) innate immunotherapy such as TLRs agonists, (4) chemo- or radiotherapy, (5) drugs able to increase antigen presentation such as the COX-2, JAK1/2 inhibitor or the MEK inhibitor cobimetinib, and (6) targeted therapy (anti-HER2, anti-VEGFR2) [69, 114, 115].

From a clinician perspective, the use of immunotherapies in recent clinical trials gave us the opportunity to contribute to a paradigmatic shift in the treatment of GI cancers. We are glad to observe highly pretreated patients experiencing a dramatic clinical benefit after treatment start, with symptoms relief, long lasting disease stabilization, and an overall manageable safety profile. We are really feeling a revolution in the daily life of our patients. Every day we ask questions about future availability of clinical trials involving immunotherapeutic agents for GI cancers from our new and historical patients. We strongly believe that further steps of drugs development such as larger phases II and III clinical trials are warranted in order to answer unsolved question and to establish the efficacy of immunotherapeutic agents. A wide international involvement of experienced centers in the next clinical trials will break a potential unequal distribution of immunotherapeutic resources.

Conflicts of Interest

The authors declare that there are no conflicts of interest regarding the publication of this paper.

Authors' Contributions

Letizia Procaccio and Marta Schirripa equally contributed as first authors. Vittorina Zagonel and Sara Lonardi equally contributed as senior authors.

References

1. Toomey P. G., Vohra N. A., Ghansah T., Sarnaik A. A., Pilon-Thomas S. A. Immunotherapy for gastrointestinal malignancies. Cancer Control. 2013;20(1):32–42. [PMC free article] [PubMed]
2. Pernot S., Terme M., Voron T., et al. Colorectal cancer and immunity: what we know and perspectives. World Journal of Gastroenterology. 2014;20(14):3738–3750. doi: 10.3748/wjg.v20.i14.3738. [PMC free article] [PubMed] [Cross Ref]
3. Vesely M. D., Schreiber R. D. Cancer immunoediting: Antigens, mechanisms, and implications to cancer immunotherapy. Annals of the New York Academy of Sciences. 2013;1284(1):1–5. doi: 10.1111/nyas.12105. [PMC free article] [PubMed] [Cross Ref]
4. Zumwalt T. J., Goel A. Immunotherapy of Metastatic Colorectal Cancer: Prevailing Challenges and New Perspectives. Current Colorectal Cancer Reports. 2015;11(3):125–140. doi: 10.1007/s11888-015-0269-2. [PMC free article] [PubMed] [Cross Ref]
5. Jung K.-W., Won Y.-J., Kong H.-J., Oh C.-M., Lee D. H., Lee J. S. Prediction of cancer incidence and mortality in korea, 2014. Cancer Research and Treatment. 2014;46(2):124–130. doi: 10.4143/crt.2014.46.2.124. [PMC free article] [PubMed] [Cross Ref]
6. Shi L., Chen S., Yang L., Li Y. The role of PD-1 and PD-L1 in T-cell immune suppression in patients with hematological malignancies. Journal of Hematology & Oncology. 2013;6(1):p. 74. doi: 10.1186/1756-8722-6-74. [PMC free article] [PubMed] [Cross Ref]
7. Han E. Q., Li X.-L., Wang C.-R., Li T.-F., Han S.-Y. Chimeric antigen receptor-engineered T cells for cancer immunotherapy: progress and challenges. Journal of Hematology and Oncology. 2013;6, article 47 doi: 10.1186/1756-8722-6-47. [PMC free article] [PubMed] [Cross Ref]
8. Bonotto M., Garattini S. K., Basile D., et al. Immunotherapy for gastric cancers: emerging role and future perspectives. Expert Review of Clinical Pharmacology. 2017;10(6):609–619. doi: 10.1080/17512433.2017.1313113. [PubMed] [Cross Ref]
9. Croci D. O., Zacarías Fluck M. F., Rico M. J., Matar P., Rabinovich G. A., Scharovsky O. G. Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment. Cancer Immunology, Immunotherapy. 2007;56(11):1687–1700. doi: 10.1007/s00262-007-0343-y. [PubMed] [Cross Ref]
10. Chen L., Flies D. B. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nature Reviews Immunology. 2013;13(4):227–242. doi: 10.1038/nri3405. [PMC free article] [PubMed] [Cross Ref]
11. Burnet F. M. The concept of immunological surveillance. Progress in Tumor Research Home. 1970;13:1–27. [PubMed]
12. Thomas L. Cellular and humoral aspects of the hypersensitive states. Journal of Internal Medicine. 1961;170(1):p. 128. doi: 10.1111/j.0954-6820.1961.tb00220.x. [Cross Ref]
13. Gooden M. J. M., de Bock G. H., Leffers N., Daemen T., Nijman H. W. The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. British Journal of Cancer. 2011;105(1):93–103. doi: 10.1038/bjc.2011.189. [PMC free article] [PubMed] [Cross Ref]
14. Galon J., Pagès F., Marincola F. M. Cancer classification using the Immunoscore: a worldwide task force. Journal of Translational Medicine. 2012;10:p. 205. doi: 10.1186/1479-5876-10-205. [PMC free article] [PubMed] [Cross Ref]
15. Lakshmi Narendra B., Eshvendar Reddy K., Shantikumar S., Ramakrishna S. Immune system: A double-edged sword in cancer. Inflammation Research. 2013;62(9):823–834. doi: 10.1007/s00011-013-0645-9. [PubMed] [Cross Ref]
16. Schreiber R. D., Old L. J., Smyth M. J. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science. 2011;331(6024):1565–1570. doi: 10.1126/science.1203486. [PubMed] [Cross Ref]
17. Sideras K., Braat H., Kwekkeboom J., et al. Corrigendum to "Role of the immune system in pancreatic cancer progression and immune modulating treatment strategies" [Cancer Treat. Rev. 40 (2014) 513-522] Cancer Treatment Reviews. 2014;40(7):p. 892. doi: 10.1016/j.ctrv.2014.05.002. [PubMed] [Cross Ref]
18. Martínez-Bosch N., Fernández-Barrena M. G., Moreno M., et al. Galectin-1 drives pancreatic carcinogenesis through stroma remodeling and hedgehog signaling activation. Cancer Research. 2014;74(13):3512–3524. doi: 10.1158/0008-5472.CAN-13-3013. [PMC free article] [PubMed] [Cross Ref]
19. Brandacher G., Perathoner A., Ladurner R., et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clinical Cancer Research. 2006;12(4):1144–1151. doi: 10.1158/1078-0432.CCR-05-1966. [PubMed] [Cross Ref]
20. Nomi T., Sho M., Akahori T., et al. Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research. 2007;13(7):2151–2157. doi: 10.1158/1078-0432.CCR-06-2746. [PubMed] [Cross Ref]
21. Gao Q., Wang X.-Y., Qiu S.-J., et al. Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clinical Cancer Research. 2009;15(3):971–979. doi: 10.1158/1078-0432.CCR-08-1608. [PubMed] [Cross Ref]
22. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–209. [PMC free article] [PubMed]
23. Maitra A., Hruban R. H. Pancreatic cancer. Annual Review of Pathology: Mechanisms of Disease. 2008;3:157–188. doi: 10.1146/annurev.pathmechdis.3.121806.154305. [PMC free article] [PubMed] [Cross Ref]
24. Coussens L. M., Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–867. doi: 10.1038/nature01322. [PMC free article] [PubMed] [Cross Ref]
25. Ghiringhelli F., Ménard C., Terme M., et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-β-dependent manner. Journal of Experimental Medicine. 2005;202(8):1075–1085. doi: 10.1084/jem.20051511. [PMC free article] [PubMed] [Cross Ref]
26. Waghray M., Yalamanchili M., Magliano M. P. D., Simeone D. M. Deciphering the role of stroma in pancreatic cancer. Current Opinion in Gastroenterology. 2013;29(5):537–543. doi: 10.1097/MOG.0b013e328363affe. [PMC free article] [PubMed] [Cross Ref]
27. Haqq J., Howells L. M., Garcea G., Metcalfe M. S., Steward W. P., Dennison A. R. Pancreatic stellate cells and pancreas cancer: Current perspectives and future strategies. European Journal of Cancer. 2014;50(15):2570–2582. doi: 10.1016/j.ejca.2014.06.021. [PubMed] [Cross Ref]
28. Mueller M. M., Fusenig N. E. Friends or foes—bipolar effects of the tumour stroma in cancer. Nature Reviews Cancer. 2004;4(11):839–849. doi: 10.1038/nrc1477. [PubMed] [Cross Ref]
29. Herrera M., Herrera A., Domínguez G., et al. Cancer-associated fibroblast and M2 macrophage markers together predict outcome in colorectal cancer patients. Cancer Science. 2013;104(4):437–444. doi: 10.1111/cas.12096. [PubMed] [Cross Ref]
30. Vonderheide R. H., Bajor D. L., Winograd R., Evans R. A., Bayne L. J., Beatty G. L. CD40 immunotherapy for pancreatic cancer. Cancer Immunology, Immunotherapy. 2013;62(5):949–954. doi: 10.1007/s00262-013-1427-5. [PMC free article] [PubMed] [Cross Ref]
31. Moran A. E., Kovacsovics-Bankowski M., Weinberg A. D. The TNFRs OX40, 4-1BB, and CD40 as targets for cancer immunotherapy. Current Opinion in Immunology. 2013;25(2):230–237. doi: 10.1016/j.coi.2013.01.004. [PMC free article] [PubMed] [Cross Ref]
32. Yamamoto T., Yanagimoto H., Satoi S., et al. Circulating CD4 +CD25 + regulatory T cells in patients with pancreatic cancer. Pancreas. 2012;41(3):409–415. doi: 10.1097/MPA.0b013e3182373a66. [PubMed] [Cross Ref]
33. Dunn G. P., Old L. J., Schreiber R. D. The immunobiology of cancer immunosurveillance and immunoediting. Immunity and Immunoediting. 2004;21(2):137–148. doi: 10.1016/j.immuni.2004.07.017. [PubMed] [Cross Ref]
34. Topalian S. L., Drake C. G., Pardoll D. M. Targeting the PD-1/B7-H1(PD-L1) pathway to activate anti-tumor immunity. Current Opinion in Immunology. 2012;24(2):207–212. doi: 10.1016/j.coi.2011.12.009. [PMC free article] [PubMed] [Cross Ref]
35. Blank C., Gajewski T. F., Mackensen A. Interaction of PD-L1 on tumor cells with PD-1 on tumor-specific T cells as a mechanism of immune evasion: Implications for tumor immunotherapy. Cancer Immunology, Immunotherapy. 2005;54(4):307–314. doi: 10.1007/s00262-004-0593-x. [PubMed] [Cross Ref]
36. Markman J. L., Shiao S. L. Impact of the immune system and immunotherapy in colorectal cancer. Journal of Gastrointestinal Oncology. 2015;6(2):208–223. [PMC free article] [PubMed]
37. Hanna M. G., Jr., Hoover H. C., Jr., Vermorken J. B., Harris J. E., Pinedo H. M. Adjuvant active specific immunotherapy of stage II and stage III colon cancer with an autologous tumor cell vaccine: First randomized phase III trials show promise. Vaccine. 2001;19(17-19):2576–2582. doi: 10.1016/S0264-410X(00)00485-0. [PubMed] [Cross Ref]
38. Derouazi M., Di Berardino-Besson W., Belnoue E., et al. Novel cell-penetrating peptide-based vaccine induces robust CD4+ and CD8+ T cell-mediated antitumor immunity. Cancer Research. 2015;75(15):3020–3031. doi: 10.1158/0008-5472.CAN-14-3017. [PubMed] [Cross Ref]
39. Robert C., Schachter J., Long G. V., et al. Pembrolizumab versus ipilimumab in advanced melanoma. The New England Journal of Medicine. 2015;372(26):2521–2532. doi: 10.1056/nejmoa1503093. [PubMed] [Cross Ref]
40. Valsecchi M. E. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. New England Journal of Medicine. 2015;373(13):p. 1270. doi: 10.1056/NEJMc1509660. [PubMed] [Cross Ref]
41. Aparicio T. PD-1 blockade in tumors with mismatch-repair deficiency: Le DT (2015) N Engl J Med May 30. Colon and Rectum. 2015;9(3):182–184. doi: 10.1007/s11725-015-0588-4. [Cross Ref]
42. Besser M. J., Shapira-Frommer R., Treves A. J., et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clinical Cancer Research. 2010;16(9):2646–2655. doi: 10.1158/1078-0432.CCR-10-0041. [PubMed] [Cross Ref]
43. Rosenberg S. A. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clinical Cancer Research. 2011;17(13):4550–4557. [PMC free article] [PubMed]
44. Amedei A., Niccolai E., D'Elios M. M. T cells and adoptive immunotherapy: recent developments and future prospects in gastrointestinal oncology. Clinical and Developmental Immunology. 2011;2011:17. doi: 10.1155/2011/320571.320571 [PMC free article] [PubMed] [Cross Ref]
45. Morgan R. A., Yang J. C., Kitano M., Dudley M. E., Laurencot C. M., Rosenberg S. A. Case report of a serious adverse event following the administration of t cells transduced with a chimeric antigen receptor recognizing ERBB2. Molecular Therapy. 2010;18(4):843–851. doi: 10.1038/mt.2010.24. [PMC free article] [PubMed] [Cross Ref]
46. Muro K., Chung H. C., Shankaran V., et al. Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1b trial. The Lancet Oncology. 2016;17(6):717–726. doi: 10.1016/S1470-2045(16)00175-3. [PubMed] [Cross Ref]
49. Doi T., Piha-Paul S. A., Jalal S. I., et al. Updated results for the advanced esophageal carcinoma cohort of the phase Ib KEYNOTE-028 study of pembrolizumab (MK-3475) Journal of Clinical Oncology. 2016;34(4):7. doi: 10.1200/jco.2016.34.4_suppl.7. [Cross Ref]
50. Kang Y., Satoh T., Ryu M., et al. Nivolumab (ONO-4538/BMS-936558) as salvage treatment after second or later-line chemotherapy for advanced gastric or gastro-esophageal junction cancer (AGC): a double-blinded, randomized, phase III trial. Journal of Clinical Oncology. 2017;35(4) abstract 2:2. doi: 10.1200/JCO.2017.35.4_suppl.2. [Cross Ref]
51. Kojima T., Hara H., Yamaguchi K., et al. Phase II study of nivolumab (ONO-4538/BMS-936558) in patients with esophageal cancer: preliminary report of overall survival. Journal of Clinical Oncology. 2016;34(supplement 4):TPS175. doi: 10.1200/jco.2016.34.4_suppl.tps175. [Cross Ref]
52. Oh D., Lockhart A. C., Wong D. J., et al. Avelumab (MSB0010718C), an anti-PD-L1 antibody, as a third-line treatment in patients with advanced gastric or gastroesophageal junction cancer: a phase Ib JAVELIN Solid Tumor trial. Journal of Clinical Oncology. 2016;34(supplement 4) doi: 10.1200/jco.2016.34.4_suppl.tps188. [Cross Ref]
53. https://clinicaltrials.gov/ct2/show/NCT02625610, ?term=javelin+gastricrank=2.
54. https://clinicaltrials.gov/ct2/show/NCT02625623, ?term=javelin+gastricrank=1.
55. Ralph C., Elkord E., Burt D. J., et al. Modulation of lymphocyte regulation for cancer therapy: a phase II trial of tremelimumab in advanced gastric and esophageal adenocarcinoma. Clinical Cancer Research. 2010;16(5):1662–1672. doi: 10.1158/1078-0432.ccr-09-2870. [PubMed] [Cross Ref]
56. Moehler M. H., Cho J. Y., Kim Y. H., et al. A randomized, open-label, two-arm phase II trial comparing the efficacy of sequential ipilimumab (ipi) versus best supportive care (BSC) following first-line (1L) chemotherapy in patients with unresectable, locally advanced/metastatic (A/M) gastric or gastro-esophageal junction (G/GEJ) cancer. Journal of Clinical Oncology. 2016;34
57. Janjigian Y. Y., Bendell J. C., Calvo E., et al. CheckMate-032: Phase I/II, open-label study of safety and activity of nivolumab (nivo) alone or with ipilimumab (ipi) in advanced and metastatic (A/M) gastric cancer (GC) Journal of Clinical Oncology. 2016;34
58. Knudsen E., Vail P., Balaji U., et al. Stratification of pancreatic ductal adenocarcinoma: combinatorial genetic, stromal, and immunological markers. Clinical Cancer Research. 2017 doi: 10.1158/1078-0432.CCR-17-0162. [PubMed] [Cross Ref]
59. Hoover H. C., Jr., Peters L. C., Brandhorst J. S., Hanna M. G., Jr. Therapy of spontaneous metastases with an autologous tumor vaccine in a guinea pig model. Journal of Surgical Research. 1981;30(4):409–415. doi: 10.1016/0022-4804(81)90180-3. [PubMed] [Cross Ref]
60. Hoover H. C., Jr., Brandhorst J. S., Peters L. C., et al. Adjuvant active specific immunotherapy for human colorectal cancer: 6.5-year median follow-up of a phase III prospectively randomized trial. Journal of Clinical Oncology. 1993;11(3):390–399. doi: 10.1200/JCO.1993.11.3.390. [PubMed] [Cross Ref]
61. Harris J. E., Ryan L., Hoover H. C., Jr., et al. Adjuvant active specific immunotherapy for stage II and III colon cancer with an autologous tumor cell vaccine: Eastern Cooperative Oncology Group study E5283. Journal of Clinical Oncology. 2000;18(1):148–157. doi: 10.1200/JCO.2000.18.1.148. [PubMed] [Cross Ref]
62. Vermorken J. B., Claessen A. M. E., Van Tinteren H., et al. Active specific immunotherapy for stage II and stage III human colon cancer: A randomised trial. Lancet. 1999;353(9150):345–350. doi: 10.1016/S0140-6736(98)07186-4. [PubMed] [Cross Ref]
63. Liebrich W., Schlag P., Manasterski M., et al. In vitro and clinical characterisation of a newcastle disease virus-modified autologous tumour cell vaccine for treatment of colorectal cancer patients. European Journal of Cancer and Clinical Oncology. 1991;27(6):703–710. doi: 10.1016/0277-5379(91)90170-I. [PubMed] [Cross Ref]
64. Schulze T., Kemmner W., Weitz J., Wernecke K.-D., Schirrmacher V., Schlag P. M. Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: Results of a prospective randomized trial. Cancer Immunology, Immunotherapy. 2009;58(1):61–69. doi: 10.1007/s00262-008-0526-1. [PubMed] [Cross Ref]
65. Llosa N. J., Cruise M., Tam A., et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discovery. 2015;5(1):43–51. doi: 10.1158/2159-8290.CD-14-0863. [PMC free article] [PubMed] [Cross Ref]
68. Le D. T. Programmed death-1 blockade in mismatch repair deficient colorectal cancer. Journal of Clinical Oncology. 2016;34(29):3502–3510. doi: 10.1200/JCO.2015.61.8983. [Cross Ref]
69. Overman M., Kopetz S., Lonardi S., et al. Nivolumab ± ipilimumab treatment (Tx) efficacy, safety, and biomarkers in patients (Pts) with metastatic colorectal cancer (mCRC) with and without high microsatellite instability (MSI-H): results from the CheckMate-142 study. Annals of Oncology. 2016;34(supplementry 6) doi: 10.1093/annonc/mdw370.27. [Cross Ref]
70. Bendell J. C., Kim T. W., Goh B. C., et al. Clinical activity and safety of cobimetinib (cobi) and atezolizumab in colorectal cancer (CRC) Journal of Clinical Oncology. 2016;34
71. Overman M. J., Lonardi S., Leone F., et al. Nivolumab in patients with DNA mismatch repair deficient/microsatellite instability high metastatic colorectal cancer: update from CheckMate 142. Journal of Clinical Oncology. 2017;35(supplementry 4):519. doi: 10.1200/JCO.2017.35.4_suppl.519. [Cross Ref]
72. Ebert P. J. R., Cheung J., Yang Y., et al. MAP Kinase Inhibition Promotes T Cell and Anti-tumor Activity in Combination with PD-L1 Checkpoint Blockade. Immunity. 2016;44(3):609–621. doi: 10.1016/j.immuni.2016.01.024. [PubMed] [Cross Ref]
73. Llovet J. M., Sala M., Castells L., et al. Randomized controlled trial of interferon treatment for advanced hepatocellular carcinoma. Hepatology. 2000;31(1):54–58. doi: 10.1002/hep.510310111. [PubMed] [Cross Ref]
74. Chen L.-T., Chen M.-F., Li L.-A., et al. Long-term results of a randomized, observation-controlled, phase III Trial of Adjuvant Interferon alfa-2b in hepatocellular carcinoma after curative resection. Annals of Surgery. 2012;255(1):8–17. doi: 10.1097/SLA.0b013e3182363ff9. [PubMed] [Cross Ref]
75. Butterfield L. H., Ribas A., Meng W. S., et al. T-cell responses to HLA-A∗0201 immunodominant peptides derived from alpha-fetoprotein in patients with hepatocellular cancer. Clinical Cancer Research. 2003;9:p. 5902. [PubMed]
76. Butterfield L. H., Ribas A., Dissette V. B., et al. A phase I/II trial testing immunization of hepatocellular carcinoma patients with dendritic cells pulsed with four α-fetoprotein peptides. Clinical Cancer Research. 2006;12(9):2817–2825. doi: 10.1158/1078-0432.CCR-05-2856. [PubMed] [Cross Ref]
77. Sawada Y., Yoshikawa T., Nobuoka D., et al. Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clinical Cancer Research. 2012;18(13):3686–3696. doi: 10.1158/1078-0432.ccr-11-3044. [PubMed] [Cross Ref]
78. Sawada Y., Yoshikawa T., Ofuji K., et al. Phase II study of the GPC3-derived peptide vaccine as an adjuvant therapy for hepatocellular carcinoma patients. OncoImmunology. 2016;5(5) doi: 10.1080/2162402X.2015.1129483.e1129483 [PMC free article] [PubMed] [Cross Ref]
79. Greten T. F., Forner A., Korangy F., et al. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer. 2010;10, article no. 209 doi: 10.1186/1471-2407-10-209. [PMC free article] [PubMed] [Cross Ref]
80. Lan Y.-H., Li Y.-G., Liang Z.-W., et al. A DNA vaccine against chimeric AFP enhanced by HSP70 suppresses growth of hepatocellular carcinoma. Cancer Immunology, Immunotherapy. 2007;56(7):1009–1016. doi: 10.1007/s00262-006-0254-3. [PubMed] [Cross Ref]
81. Li S. Q., Lin J., Qi C. Y., et al. GPC3 DNA vaccine elicits potent cellular antitumor immunity against HCC in mice. Hepatogastroenterology. 2014;61(130):278–84. [PubMed]
82. Butterfield L. H., Economou J. S., Gamblin T. C., Geller D. A. Alpha fetoprotein DNA prime and adenovirus boost immunization of two hepatocellular cancer patients. Journal of Translational Medicine. 2014;12(1, article no. 86) doi: 10.1186/1479-5876-12-86. [PMC free article] [PubMed] [Cross Ref]
83. Lee W. C., Wang H. C., Hung C. F., Huang P. F., Lia C. R., Chen M. F. Vaccination of advanced hepatocellular carcinoma patients with tumor lysate-pulsed dendritic cells: a clinical trial. Journal of Immunotherapy. 2005;28(5):496–504. doi: 10.1097/01.cji.0000171291.72039.e2. [PubMed] [Cross Ref]
84. Palmer D. H., Midgley R. S., Mirza N., et al. A phase II study of adoptive immunotherapy using dendritic cells pulsed with tumor lysate in patients with hepatocellular carcinoma. Hepatology. 2009;49(1):124–132. doi: 10.1002/hep.22626. [PubMed] [Cross Ref]
85. El Ansary M., Mogawer S., Elhamid S. A., et al. Immunotherapy by autologous dendritic cell vaccine in patients with advanced HCC. Journal of Cancer Research and Clinical Oncology. 2013;139(1):39–48. doi: 10.1007/s00432-012-1298-8. [PMC free article] [PubMed] [Cross Ref]
86. Tada F., Abe M., Hirooka M., et al. Phase I/II study of immunotherapy using tumor antigen-pulsed dendritic cells in patients with hepatocellular carcinoma. International Journal of Oncology. 2012;41(5):1601–1609. doi: 10.3892/ijo.2012.1626. [PMC free article] [PubMed] [Cross Ref]
87. Sangro B., Gomez-Martin C., de la Mata M. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. Journal of Hepatology. 2013;59(1):81–88. doi: 10.1016/j.jhep.2013.02.022. [PubMed] [Cross Ref]
88. Melero I., Sangro B., Yau T. C., et al. Nivolumab dose escalation and expansion in patients with advanced hepatocellular carcinoma (HCC): the CheckMate 040 study. Journal of Clinical Oncology. 2017;35(supplementry 4):226. doi: 10.1200/JCO.2017.35.4_suppl.226. [Cross Ref]
89. Llovet J. M., Ricci S., Mazzaferro V., et al. Sorafenib in Advanced Hepatocellular Carcinoma. New England Journal of Medicine. 2008;359(23):2497–2499. doi: 10.1056/NEJMc081780. [PubMed] [Cross Ref]
90. Bruix J., Raou J.-L., Sherman M., et al. Efficacy and safety of sorafenib in patients with advanced hepatocellular carcinoma: subanalyses of a phase III trial. Journal of Hepatology. 2012;57(4):821–829. doi: 10.1016/j.jhep.2012.06.014. [PubMed] [Cross Ref]
91. Sangro B., Park J.-W., Dela Cruz C. M., et al. A randomized, multicenter, phase 3 study of nivolumab vs sorafenib as first-line treatment in patients (pts) with advanced hepatocellular carcinoma (HCC): CheckMate-459. Journal of Clinical Oncology. 2016;34
92. Segal N., Hamid O., Hwu W., et al. A phase I multi-arm dose-expansion study of the anti-programmed cell death-ligand-1 (PD-L1) antibody MEDI4736: preliminary data. Annals of Oncology. 2014;25(supplementry 4) doi: 10.1093/annonc/mdu342.11. [Cross Ref]
93. Lovitch S. B., Rodig S. J. The Role of Surgical Pathology in Guiding Cancer Immunotherapy. Annual Review of Pathology: Mechanisms of Disease. 2016;11:313–341. doi: 10.1146/annurev-pathol-012615-044125. [PubMed] [Cross Ref]
94. Gibney G. T., Weiner L. M., Atkins M. B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. The Lancet Oncology. 2016;17(12):e542–e551. doi: 10.1016/S1470-2045(16)30406-5. [PubMed] [Cross Ref]
95. Postow M. A., Callahan M. K., Wolchok J. D. Immune checkpoint blockade in cancer therapy. Journal of Clinical Oncology. 2015;33(17):1974–1982. doi: 10.1200/jco.2014.59.4358. [PMC free article] [PubMed] [Cross Ref]
96. Patel S. P., Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Molecular Cancer Therapeutics. 2015;14(4):847–856. doi: 10.1158/1535-7163.mct-14-0983. [PubMed] [Cross Ref]
97. Herbst R. S., Soria J. C., Kowanetz M. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515(7528):563–567. doi: 10.1038/nature14011. [PMC free article] [PubMed] [Cross Ref]
98. Fassan M., Baffa R., Kiss A. Advanced precancerous lesions within the GI tract: The molecular background. Best Practice and Research: Clinical Gastroenterology. 2013;27(2):159–169. doi: 10.1016/j.bpg.2013.03.009. [PubMed] [Cross Ref]
99. Eric B. Next-generation sequencing and immunotherapy biomarkers: A medical oncology perspective. Archives of Pathology and Laboratory Medicine. 2016;140(3):245–248. doi: 10.5858/arpa.2015-0287-SA. [PubMed] [Cross Ref]
100. Umar A. Revised bethesda guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. Journal of the National Cancer Institute. 2004;96(18):1403–1404. doi: 10.1093/jnci/djh281. [PubMed] [Cross Ref]
101. Khagi Y., Kurzrock R., Patel S. P. Next generation predictive biomarkers for immune checkpoint inhibition. Cancer and Metastasis Reviews. 2016:1–12. doi: 10.1007/s10555-016-9652-y. [PMC free article] [PubMed] [Cross Ref]
102. Gajewski T. F., Schreiber H., Fu Y. X. Innate and adaptive immune cells in the tumor microenvironment. Nature Immunology. 2013;14:1014–1022. doi: 10.1038/ni.2703. [PMC free article] [PubMed] [Cross Ref]
103. Solinas C., Pusole G., Demurtas L., et al. Tumor infiltrating lymphocytes in gastrointestinal tumors: controversies and future clinical implications. Critical Reviews in Oncology/Hematology. 2017;110:106–116. doi: 10.1016/j.critrevonc.2016.11.016. [PubMed] [Cross Ref]
104. Ferris R. L., Galon J. Additional support for the introduction of immune cell quantification in colorectal cancer classification. Journal of the National Cancer Institute. 2016;108(8) doi: 10.1093/jnci/djw033.djw033 [PMC free article] [PubMed] [Cross Ref]
105. Galon J., Mlecnik B., Bindea G., et al. Towards the introduction of the ‘Immunoscore’ in the classification of malignant tumours. Journal of Pathology. 2014;232(2):199–209. doi: 10.1002/path.4287. [PMC free article] [PubMed] [Cross Ref]
106. Tian S., Roepman P., Popovici V., et al. A robust genomic signature for the detection of colorectal cancer patients with microsatellite instability phenotype and high mutation frequency. Journal of Pathology. 2012;228(4):586–595. doi: 10.1002/path.4092. [PMC free article] [PubMed] [Cross Ref]
107. Mlecnik B., Bindea G., Angell H. K., et al. Integrative Analyses of Colorectal Cancer Show Immunoscore Is a Stronger Predictor of Patient Survival Than Microsatellite Instability. Immunity. 2016;44(3):698–711. doi: 10.1016/j.immuni.2016.02.025. [PubMed] [Cross Ref]
108. Zhao H., Thienpont B., Yesilyurt B. T., et al. Mismatch repair deficiency endows tumors with a unique mutation signature and sensitivity to DNA double-strand breaks. eLife. 2014;3(2014):1–26. doi: 10.7554/eLife.02725.e02725 [PMC free article] [PubMed] [Cross Ref]
109. Chen K.-H., Yuan C.-T., Tseng L.-H., Shun C.-T., Yeh K.-H. Case report: Mismatch repair proficiency and microsatellite stability in gastric cancer may not predict programmed death-1 blockade resistance. Journal of Hematology and Oncology. 2016;9(1, article no. 29) doi: 10.1186/s13045-016-0259-0. [PMC free article] [PubMed] [Cross Ref]
110. Vétizou M., Pitt J. M., Daillère R., et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi: 10.1126/science.aad1329. [PMC free article] [PubMed] [Cross Ref]
111. Lote H., Cafferkey C., Chau I. PD-1 and PD-L1 blockade in gastrointestinal malignancies. Cancer Treatment Reviews. 2015;41(10):893–903. doi: 10.1016/j.ctrv.2015.09.004. [PubMed] [Cross Ref]
112. Wolchok J. D., Hoos A., O'Day S., et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clinical Cancer Research. 2009;15(23):7412–7420. doi: 10.1158/1078-0432.CCR-09-1624. [PubMed] [Cross Ref]
113. Ciccarese C., Alfieri S., Santoni M., et al. New toxicity profile for novel immunotherapy agents: Focus on immune-checkpoint inhibitors. Expert Opinion on Drug Metabolism and Toxicology. 2016;12(1):57–74. doi: 10.1517/17425255.2016.1120287. [PubMed] [Cross Ref]
114. Robert C., Thomas L., Bondarenko I. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. The New England Journal of Medicine. 2011;364(26):2517–2526. doi: 10.1056/NEJMoa1104621. [PubMed] [Cross Ref]
115. Hodi F. S., Chesney J., Pavlick A. C., et al. Combined nivolumab and ipilimumab versus ipilimumab alone in patients with advanced melanoma: 2-year overall survival outcomes in a multicentre, randomised, controlled, phase 2 trial. The Lancet Oncology. 2016;17(11):1558–1568. doi: 10.1016/S1470-2045(16)30366-7. [PubMed] [Cross Ref]

Articles from BioMed Research International are provided here courtesy of Hindawi

Current Status and Perspective of Immunotherapy in Gastrointestinal Cancers

J Cancer. 2016; 7(12): 1599–1604.
Published online 2016 Jul 18. doi:  10.7150/jca.16208
PMCID: PMC5039380

Current Status and Perspective of Immunotherapy in Gastrointestinal Cancers

Abstract

Cancer immunotherapy is at dawn of the Renaissance after the Medieval Dark Ages. Recent advances of understanding tumor immunology and molecular drug development are leading us to the epoch of cancer immunotherapy. Some types of immunotherapy have shown to provide survival benefit for patients with solid tumors such as malignant melanoma, renal cell carcinoma, or non-small cell lung cancer. Several studies have suggested that immune checkpoint inhibition might be effective in some patients with gastrointestinal cancers. However, the era of cancer immunotherapy in gastrointestinal cancers is still in an inchoate stage. Here we briefly review the current status and perspective of immunotherapeutic approaches in patients with gastrointestinal cancers.

Conclusion

Cancer immunotherapy has shown promising results in various types of cancers. Especially immune checkpoint inhibitors are leading a recent renaissance of immune-mediated anticancer treatments. Early studies have suggested that immune checkpoint inhibition might also be effective in some patients with gastrointestinal cancers. However, the era of cancer immunotherapy in gastrointestinal cancers is in an inchoate stage. The immune system and immunosuppressive mechanisms surrounding GC or CRC are still obscure compared to malignant melanoma, renal cell carcinoma, or NSCLC. To increase efficacy of cancer immunotherapy in gastrointestinal cancers, we need to build more profound understanding of tumor immune system. Knowledge of potential relationships between tumor cells and their microenvironment is also essential in gastrointestinal malignancies.

References

1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M. et al. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN2012. Int J Cancer. 2015;136:E359–86. [PubMed]
2. Jung KW, Won YJ, Kong HJ, Oh CM, Lee DH, Lee JS. Prediction of cancer incidence and mortality in Korea, 2014. Cancer Res Treat. 2014;46:124–30. [PMC free article] [PubMed]
3. Slankard-Chahinian M, Holland JF, Gordon RE, Becker J, Ohnuma T. Adoptive autoimmunotherapy. Cytotoxic effect of an autologous long-term T-cell line on malignant melanoma. Cancer. 1984;53:1066–72. [PubMed]
4. Parkinson DR, Abrams JS, Wiernik PH, Rayner AA, Margolin KA, Van Echo DA. et al. Interleukin-2 therapy in patients with metastatic malignant melanoma: a phase II study. J Clin Oncol. 1990;8:1650–6. [PubMed]
5. Flaherty LE, Atkins M, Sosman J, Weiss G, Clark JI, Margolin K. et al. Outpatient biochemotherapy with interleukin-2 and interferon alfa-2b in patients with metastatic malignant melanoma: results of two phase II cytokine working group trials. J Clin Oncol. 2001;19:3194–202. [PubMed]
6. Bar MH, Sznol M, Atkins MB, Ciobanu N, Micetich KC, Boldt DH. et al. Metastatic malignant melanoma treated with combined bolus and continuous infusion interleukin-2 and lymphokine-activated killer cells. J Clin Oncol. 1990;8:1138–47. [PubMed]
7. Starr P. Encouraging results for pembrolizumab in head and neck cancer. Am Health Drug Benefits. 2015;8:16. [PMC free article] [PubMed]
8. Prall F, Dührkop T, Weirich V, Ostwald C, Lenz P, Nizze H, Barten M. Prognostic role of CD8+ tumor-infiltrating lymphocytes in stage III colorectal cancer with and without microsatellite instability. Hum Pathol. 2004;35:808–16. [PubMed]
9. Karlsson M, Marits P, Dahl K, Dagöö T, Enerbäck S, Thörn M, Winqvist O. Pilot study of sentinel-node-based adoptive immunotherapy in advanced colorectal cancer. Ann Surg Oncol. 2010;17:1747–57. [PMC free article] [PubMed]
10. Jiang J, Xu N, Wu C, Deng H, Lu M, Li M. et al. Treatment of advanced gastric cancer by chemotherapy combined with autologous cytokine-induced killer cells. Anticancer Res. 2006;26:2237–42. [PubMed]
11. Kim JH, Lee Y, Bae Y-S, Kim WS, Kim K, Im HY. et al. Phase I/II study of immunotherapy using autologous tumor lysate-pulsed dendritic cells in patients with metastatic renal cell carcinoma. Clin Immunol. 2007;125:257–67. [PubMed]
12. Testori A, Richards J, Whitman E, Mann GB, Lutzky J, Camacho L. et al. Phase III comparison of vitespen, an autologous tumor-derived heat shock protein gp96 peptide complex vaccine, with physician's choice of treatment for stage IV melanoma: The C-100-21 Study Group. J Clin Oncol. 2008;26:955–62. [PubMed]
13. Um S-J, Choi YJ, Shin H-J, Son CH, Park Y-S, Roh MS. et al. Phase I study of autologous dendritic cell tumor vaccine in patients with non-small cell lung cancer. Lung Cancer. 2010;70:188–94. [PubMed]
14. Hanna Jr. MG, Hoover Jr. HC, Vermorken JB, Harris JE, Pinedo HM. Adjuvant active specific immunotherapy of stage II and stage III colon cancer with an autologous tumor cell vaccine: first randomized phase III trials show promise. Vaccine. 2001;19:2576–82. [PubMed]
15. Conry RM, Curiel DT, Strong TV, Moore SE, Allen KO, Barlow DL. et al. Safety and immunogenicity of a DNA vaccine encoding carcinoembryonic antigen and hepatitis B surface antigen in colorectal carcinoma patients. Clin Cancer Res. 2002;8:2782–7. [PubMed]
16. Mazzaferro V, Coppa J, Carrabba MG, Rivoltini L, Schiavo M, Regalia E. et al. Vaccination with autologous tumor-derived heat-shock protein Gp96 after liver resection for metastatic colorectal cancer. Clin Cancer Res. 2003;9:3235–45. [PubMed]
17. Lasky JL, Panosyan EH, Plant A, Davidson T, Yong WH, Prins RM. et al. Autologous tumor lysate-pulsed dendritic cell immunotherapy for pediatric patients with newly diagnosed or recurrent high-grade gliomas. Anticancer Res. 2013;33:2047–56. [PMC free article] [PubMed]
18. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF. et al. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–22. [PubMed]
19. Sadanaga N, Nagashima H, Mashino K, Tahara K, Yamaguchi H, Ohta M. et al. Dendritic cell vaccination with MAGE peptide is a novel therapeutic approach for gastrointestinal carcinomas. Clin Cancer Res. 2001;7:2277–84. [PubMed]
20. Kono K, Takahashi A, Sugai H, Fujii H, Choudhury AR, Kiessling R. et al. Dendritic cells pulsed with HER-2/neu-derived peptides can induce specific T-cell responses in patients with gastric cancer. Clin Cancer Res. 2002;8:3394–400. [PubMed]
21. Czerniecki BJ, Koski GK, Koldovsky U, Xu S, Cohen PA, Mick R. et al. Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res. 2007;67:1842–52. [PubMed]
22. Kim JH, Kang TH, Noh KH, Kim S-H, Lee Y-H, Kim KW. et al. Enhancement of DC vaccine potency by activating the PI3K/AKT pathway with a small interfering RNA targeting PTEN. Immunol Lett. 2010;134:47–54. [PubMed]
23. Ojima T, Iwahashi M, Nakamura M, Matsuda K, Nakamori M, Ueda K. et al. Benefits of gene transduction of granulocyte macrophage colony-stimulating factor in cancer vaccine using genetically modified dendritic cells. Int J Oncol. 2007;31:931–9. [PubMed]
24. Morse MA, Niedzwiecki D, Marshall JL, Garrett C, Chang DZ, Aklilu M. et al. A randomized phase II study of immunization with dendritic cells modified with poxvectors encoding CEA and MUC1 compared with the same poxvectors plus GM-CSF for resected metastatic colorectal cancer. Ann Surg. 2016;258:879–86. [PMC free article] [PubMed]
25. Barth RJ, Fischer DA, Wallace PK, Channon JY, Noelle RJ, Gui J, Ernstoff MS. A randomized trial of ex vivo CD40L activation of a dendritic cell vaccine in colorectal cancer patients: tumor-specific immune responses are associated with improved survival. Clin Cancer Res. 2010;16:5548–56. [PMC free article] [PubMed]
26. Zhang L, Zhu W, Li J, Yang X, Ren Y, Niu J, Pang Y. Clinical outcome of immunotherapy with dendritic cell vaccine and cytokine-induced killer cell therapy in hepatobiliary and pancreatic cancer. Mol Clin Oncol. 2016;4:129–33. [PMC free article] [PubMed]
27. Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C. et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364:2517–26. [PubMed]
28. O'Mahony D, Morris JC, Quinn C, Gao W, Wilson WH, Gause B. et al. A pilot study of CTLA-4 blockade after cancer vaccine failure in patients with advanced malignancy. Clin Cancer Res. 2007;13:958–64. [PubMed]
29. Chung KY, Gore I, Fong L, Venook A, Beck SB, Dorazio P. et al. Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Oncol. 2010;28:3485–90. [PubMed]
30. Ralph C, Elkord E, Burt DJ, O'Dwyer JF, Austin EB, Stern PL. et al. Modulation of lymphocyte regulation for cancer therapy: a phase II trial of tremelimumab in advanced gastric and esophageal adenocarcinoma. Clin Cancer Res. 2010;16:1662–72. [PubMed]
31. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH. et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–7. [PubMed]
32. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L. et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–32. [PubMed]
33. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY. et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387:1540–50. [PubMed]
34. Brahmer J, Reckamp KL, Baas P, Crinò L, Eberhardt WEE, Poddubskaya E. et al. Nivolumab versus docetaxel in advanced squamous-cell non-small cell lung cancer. N Engl J Med. 2015;373:123–35. [PMC free article] [PubMed]
35. Martin-Liberal J, Kordbacheh T, Larkin J. Safety of pembrolizumab for the treatment of melanoma. Expert Opin Drug Saf. 2015;14:957–64. [PubMed]
36. Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM. et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51. [PMC free article] [PubMed]
37. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD. et al. PD-1 blockade in tumors with mismatch repair deficiency. N Engl J Med. 2015;372:2509–20. [PMC free article] [PubMed]
38. O'Neil BH, Wallmark J, Lorente D, Elez E, Raimbourg J, Gomez-Roca C. et al. Pembrolizumab (MK-3475) for patients with advanced colorectal carcinoma (CRC): Preliminary results from KEYNOTE-028. Eur J Cancer. 2015;51(suppl 3):abstr502.
39. Bustin SA, Li S-R, Phillips S, Dorudi S. Expression of HLA Class II in colorectal cancer: evidence for enhanced immunogenicity of microsatellite-instability-positive tumours. Tumor Biol. 2001;22:294–8. [PubMed]
40. Bauer K, Michel S, Reuschenbach M, Nelius N, Doeberitz M von K, Kloor M. Dendritic cell and macrophage infiltration in microsatellite-unstable and microsatellite-stable colorectal cancer. Fam Cancer. 2011;10:557–65. [PubMed]
41. Correale P, Rotundo MS, Del Vecchio MT, Remondo C, Migali C, Ginanneschi C. et al. Regulatory (FoxP3+) T-cell tumor infiltration is a favorable prognostic factor in advanced colon cancer patients undergoing chemo or chemoimmunotherapy. J Immunother. 2010;33:435–41. [PubMed]
42. Ling A, Edin S, Wikberg ML, Öberg Å, Palmqvist R. The intratumoural subsite and relation of CD8+ and FOXP3+ T lymphocytes in colorectal cancer provide important prognostic clues. Br J Cancer. 2014;110:2551–9. [PMC free article] [PubMed]
43. Muro K, Bang Y, Shankaran V, Geva R, Catenacci DVT, Gupta S, Relationship between PD-L1 expression and clinical outcomes in patients with advanced gastric cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab (Pembro; MK-3475) in KEYNOTE-012. J Clin Oncol; 2015. p. 33. (suppl 3; abstr 3)
44. Le DT, Bendell JC, Calvo E, Kim JW, Ascierto PA, Sharma P. et al. Safety and activity of nivolumab monotherapy in advanced and metastatic (A/M) gastric or gastroesophageal junction cancer (GC/GEC): results from the CheckMate-032 study. J Clin Oncol. 2016;34(suppl 4S):abstr6.
45. Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ. et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–8. [PMC free article] [PubMed]
46. Calles A, Liao X, Sholl LM, Rodig SJ, Freeman GJ, Butaney M. et al. Expression of PD-1 and its ligands, PD-L1 and PD-L2, in smokers and never smokers with KRAS-mutant lung cancer. J Thorac Oncol. 2015;10:1726–35. [PubMed]
47. Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M. et al. Genomic spectra of biliary tract cancer. Nat Genet. 2015;47:1003–10. [PubMed]
48. Bang YJ, De Braud F, Piha-Paul S, Hollebecque A, Abdul Razak AR, Safety and efficacy of pembrolizumab (MK-3475) in patients (pts) with advanced biliary tract cancer: Interim results of KEYNOTE-028. Eur J Cancer. 2106;51 (suppl 3; abstr 525)
49. Doi T, Piha-Paul SA, Jalal SI, Mai-Dang H, Saraf S, Koshiji M, Updated results for the advanced esophageal carcinoma cohort of the phase Ib KEYNOTE-028 study of pembrolizumab (MK-3475) J Clin Oncol; 2016. p. 34. (suppl 4S; abstr 7)
50. Lesterhuis WJ, de Vries IJM, Aarntzen EA, de Boer A, Scharenborg NM, van de Rakt M. et al. A pilot study on the immunogenicity of dendritic cell vaccination during adjuvant oxaliplatin/capecitabine chemotherapy in colon cancer patients. Br J Cancer. 2010;103:1415–21. [PMC free article] [PubMed]
51. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34. [PubMed]
52. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, McDermott D. et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372:2006–17. [PubMed]
53. Hodi FS, Lawrence D, Lezcano C, Wu X, Zhou J, Sasada T. et al. Bevacizumab plus ipilimumab in patients with metastatic melanoma. Cancer Immunol Res. 2014;2:632–42. [PMC free article] [PubMed]
54. Nesselhut J, Marx D, Cillien N, Lange H, Regalo G, Herrmann M, Dendritic cells generated with PDL-1 checkpoint blockade for treatment of advanced pancreatic cancer. J Clin Oncol; 2015. p. 33. (suppl 3; abstr 4128)

Articles from Journal of Cancer are provided here courtesy of Ivyspring International Publisher

Plaats een reactie ...

Reageer op "Immuuntherapie met dendritische celtherapie geeft uitstekende resultaten op overall overleving en ziektevrije tijd bij darmkanker met weinig of geen zichtbare tumoren."


Gerelateerde artikelen