A Retroviral Replicating Vector Encoding Cytosine Deaminase and 5-FC Induces Immune Memory in Metastatic Colorectal Cancer Models

Kader Yagiz, Maria E Rodriguez-Aguirre, Fernando Lopez Espinoza, Tiffany T Montellano, Daniel Mendoza, Leah A Mitchell, Carlos E Ibanez, Noriyuki Kasahara, Harry E Gruber, Douglas J Jolly, Joan M Robbins, Kader Yagiz, Maria E Rodriguez-Aguirre, Fernando Lopez Espinoza, Tiffany T Montellano, Daniel Mendoza, Leah A Mitchell, Carlos E Ibanez, Noriyuki Kasahara, Harry E Gruber, Douglas J Jolly, Joan M Robbins

Abstract

Treatment of tumors with Toca 511, a gamma retroviral replicating vector encoding cytosine deaminase, followed by 5-fluorocytosine (5-FC) kills tumors by local production of 5-fluorouracil (5-FU). In brain tumor models, this treatment induces systemic anti-tumor immune responses and long-term immune-mediated survival. Phase 1 Toca 511 and Toca FC (extended-release 5-FC) clinical trials in patients with recurrent high-grade glioma show durable complete responses and promising survival data compared to historic controls. The work described herein served to expand on our earlier findings in two models of metastatic colorectal carcinoma (mCRC). Intravenous (i.v.) delivery of Toca 511 resulted in substantial tumor-selective uptake of vector into metastatic lesions. Subsequent treatment with 5-FC resulted in tumor shrinkage, improved survival, and immune memory against future rechallenge with the same CT26 CRC cell line. Similar results were seen in a brain metastasis model of mCRC. Of note, 5-FC treatment resulted in a significant decrease in myeloid-derived suppressor cells (MDSCs) in mCRC tumors in both the liver and brain. These results support the development of Toca 511 and Toca FC as a novel immunotherapeutic approach for patients with mCRC. A phase 1 study of i.v. Toca 511 and Toca FC in solid tumors, including mCRC, is currently underway (NCT02576665).

Keywords: 5-FU; MDSC; chemoimmunotherapy; durable response; immunotherapy; mCRC; retroviral vector.

Figures

Figure 1
Figure 1
Toca 511 in Combination with 5-FC Was Efficacious in a Multifocal Liver Metastasis Model of mCRC (A) Representative radiance intensities (photon/s/mouse) from the PBS and 5-FC treatment group at various time points throughout the study. After liver metastases were established with Toca 511 pre-transduced CT26-Luc cells delivered intrasplenically, each mouse was imaged by IVIS at indicated time points. 5-FC treatment (500 mg/kg, i.p., BID) was initiated on day 13 for 5 days on and 2 days off for a total of six cycles. The control group received PBS (see also Figure S1). (B) CT26-Luc bioluminescence average signal intensity at indicated time points (n = 9/group). (C) Kaplan-Meier survival analysis. After liver metastases were established with Toca 511 pre-transduced CT26 cells delivered intrasplenically, PBS or 5-FC treatment was started at day 13 post cell inoculation (n = 9/group). A total of six cycles of PBS or 5-FC treatment were administered and survival was examined out to 90 days. The table is the summary of the survival analysis out to 90 days post tumor cell implantations. (D) Cured mice from survival studies (Toca 511/CT26+5-FC; n = 5) were subcutaneously challenged with wild-type CT26 cells on day 0 (day 90 post initial tumor inoculation) at a cell dose of 5 × 105. As a control, CT26 cells were implanted into naive BALB/cJ mice (n = 9) (*p = 0.028; naive versus Toca 511/CT26+5-FC). *Statistical significance was defined as p < 0.05. Error bars represent SEM.
Figure 2
Figure 2
Upon Intrasplenic, Intravenous, or Intraportal Delivery, GFP-Expressing Vector Concentrates in Metastatic Foci in the Liver and Not in Normal Liver Tissue (A) Representative images showing luciferase (CT26-Luc cells) and GFP (Toca GFP vector) signal in livers from mice receiving intrasplenic, intravenous, or intraportal delivery of Toca GFP vector. (B) Flow cytometric analysis of excised metastatic lesions from livers collected 18 days post vector delivery. (C) CT26-Luc tumor-bearing animals received 1 day or 3 days of intravenous delivery of Toca GFP vector. Flow cytometric analysis of excised metastatic lesions from livers collected 14 or 22 days post vector delivery (n = 9/group). Numbers above the columns indicate the average GFP+ cells per group. Error bars represent SEM.
Figure 3
Figure 3
Intravenous Delivery of Toca 511, Followed by Treatment with 5-FC, Was Efficacious against Colorectal Liver Metastasis (A) Kaplan-Meier survival analysis. Mice inoculated with CT26-Luc cells intrasplenically were then administered Toca 511 i.v. for 5 consecutive days starting on day 3 post cell inoculation. PBS or 5-FC treatment started at day 12 post cell inoculation. Six cycles of PBS or 5-FC treatment were administered, and survival was examined out to 90 days. The table is the summary of the survival analysis out to 90 days post tumor cell implantations. Toca 511 and 5-FC treated animals lived significantly longer compared to Toca 511 and PBS control animals (p = 0.037). (B) Radiance intensities (photon/s/mouse) from representative animals at indicated time points. 5-FC treatment (500 mg/kg, i.p., BID) started on day 13 for 5 days on and 2 days off for 6 cycles. Control group received PBS (see also Figure S2). (C) Cured mice from survival studies (Toca 511+5-FC; n = 5) were subcutaneously challenged with wild-type CT26 cells on day 0 (day 90 post initial tumor inoculation) at a cell dose of 5 × 105. As a control, CT26 cells were implanted into naive BALB/cJ mice (n = 9) (*p = 0.04; naive versus Toca 511+5-FC). *Statistical significance was defined as p < 0.05. (D) WBCs, LYMs, NEUs, and PLTs were determined in the PBS and 5-FC treatment groups. Dotted line represents LLN for age-matched mice. Age-matched mouse LLN = WBC, 6 × 109 cells/L; LYM, 3.4 × 109 cells/L; PLT, 200 × 109 cells/L; and NEU, 3.4 × 109 cells/L. Error bars represent SEM.
Figure 4
Figure 4
Toca 511 and 5-FC Resulted in Concentrated 5-FU within Liver Metastases in a Murine Model of mCRC LC/MS measurement of 5-FC and 5-FU in plasma and tumor from mice with Toca 511 pre-transduced CT26-Luc liver metastases treated for 3.5 days with 5-FC (500 mg/kg, i.p., BID). Numbers above columns indicate average values for each group. Error bars represent SEM.
Figure 5
Figure 5
Systemic 5-FU Treatment Was Not Efficacious in a Mouse Model of Colorectal Liver Metastasis (A) Kaplan-Meier analysis showing the survival of animals with liver metastasis treated with 5-FC (500 mg/kg, i.p., BID) or 5-FU (20 mg/kg, i.p., SID) for 5 consecutive days, followed by 2 days without drug for a total of 4 cycles. Animals received no vector in this experiment. 5-FC, without the presence of vector, has no effect on survival. (B) Hematologic analysis, including values for WBCs, LYMs, and NEUs from animals treated with 5-FC or 5-FU. Animals had established CT26 liver metastases but were not given Toca 511 vector. Error bars represent SEM.
Figure 6
Figure 6
Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Liver Metastasis Model of mCRC Liver metastases treated with 5-FC visually show a decrease in MDSC populations. Mice were administered three cycles of 5-FC (500 mg/kg, i.p., BID) or PBS. (A) Immunofluorescence color staining of CD11b (myeloid cells), Gr-1 (monocytes and neutrophils), Ly6C (monocytes), and Gr-1/Ly6C (MDSCs) confirms decrease of expression with treatment of 5-FC. (B) Quantitative analysis of percentage area (n = 6) of CD11b, Gr-1, and Ly6C show decrease of MDSCs with treatment of 5-FC compared with PBS. *Statistical significance was defined as p 

Figure 7

Toca 511 and 5-FC Prolonged…

Figure 7

Toca 511 and 5-FC Prolonged Survival and Promoted Long-Term Anti-tumor Immunity in a…

Figure 7
Toca 511 and 5-FC Prolonged Survival and Promoted Long-Term Anti-tumor Immunity in a Brain Metastasis Model of mCRC (A) Kaplan-Meier analysis. BALB/cJ mice received Toca 511 pre-transduced CT26 cells intracranially and were subsequently treated with cycles of 5-FC (500 mg/kg, i.p., BID) (n = 12) or PBS (n = 5). Treatments started at day 10 post cell inoculation and were for 7 days on and 7 days off for four cycles. Survival was examined out to 150 days. (B) Long-term survivors (n = 5) were rechallenged subcutaneously at day 65 post intracranial tumor implant with parental CT26 cells, and tumor growth was monitored over time. As a control, CT26 cells were implanted into naive, age-matched, BALB/cJ mice (n = 5) (*p = 0.0001; naive versus Toca 511+5-FC). (C) Recipient athymic animals received splenocytes from either cured or naive animals and were inoculated subcutaneously with parental CT26 cells and tumor growth was monitored over time (n = 6/group) (*p = 0.04). *Statistical significance was defined as p 

Figure 8

Myeloid-Derived Suppressor Cells Decreased with…

Figure 8

Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Brain…

Figure 8
Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Brain Metastasis Model of mCRC BALB/c mice received Toca 511 pre-transduced CT26 cells intracranially and were treated with 5-FC (500 mg/kg, i.p., BID) or PBS. Treatments started at day 10 post cell inoculation and were 7 days on and 7 days off for one cycle. (A and B) The tumor (A) (*p + Gr-1+ Ly6C+ cells in the tumor (A) and spleen (B). Error bars represent SEM.
All figures (8)
Similar articles
Cited by
References
    1. Ostertag D., Amundson K.K., Lopez Espinoza F., Martin B., Buckley T., Galvão da Silva A.P., Lin A.H., Valenta D.T., Perez O.D., Ibañez C.E. Brain tumor eradication and prolonged survival from intratumoral conversion of 5-fluorocytosine to 5-fluorouracil using a nonlytic retroviral replicating vector. Neuro-oncol. 2012;14:145–159. - PMC - PubMed
    1. Raval R.R., Sharabi A.B., Walker A.J., Drake C.G., Sharma P. Tumor immunology and cancer immunotherapy: summary of the 2013 SITC primer. J. Immunother. Cancer. 2014;2:14. - PMC - PubMed
    1. Huang T.T., Hlavaty J., Ostertag D., Espinoza F.L., Martin B., Petznek H., Rodriguez-Aguirre M., Ibañez C.E., Kasahara N., Gunzburg W. Toca 511 gene transfer and 5-fluorocytosine in combination with temozolomide demonstrates synergistic therapeutic efficacy in a temozolomide-sensitive glioblastoma model. Cancer Gene Ther. 2013;20:544–551. - PubMed
    1. Huang T.T., Parab S., Burnett R., Diago O., Ostertag D., Hofman F.M., Espinoza F.L., Martin B., Ibañez C.E., Kasahara N. Intravenous administration of retroviral replicating vector, Toca 511, demonstrates therapeutic efficacy in orthotopic immune-competent mouse glioma model. Hum. Gene Ther. 2015;26:82–93. - PMC - PubMed
    1. Yagiz K., Huang T.T., Lopez Espinoza F., Mendoza D., Ibañez C.E., Gruber H.E., Jolly D.J., Robbins J.M. Toca 511 plus 5-fluorocytosine in combination with lomustine shows chemotoxic and immunotherapeutic activity with no additive toxicity in rodent glioblastoma models. Neuro-oncol. 2016;18:1390–1401. - PMC - PubMed
Show all 55 references
Associated data
Related information
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 7
Figure 7
Toca 511 and 5-FC Prolonged Survival and Promoted Long-Term Anti-tumor Immunity in a Brain Metastasis Model of mCRC (A) Kaplan-Meier analysis. BALB/cJ mice received Toca 511 pre-transduced CT26 cells intracranially and were subsequently treated with cycles of 5-FC (500 mg/kg, i.p., BID) (n = 12) or PBS (n = 5). Treatments started at day 10 post cell inoculation and were for 7 days on and 7 days off for four cycles. Survival was examined out to 150 days. (B) Long-term survivors (n = 5) were rechallenged subcutaneously at day 65 post intracranial tumor implant with parental CT26 cells, and tumor growth was monitored over time. As a control, CT26 cells were implanted into naive, age-matched, BALB/cJ mice (n = 5) (*p = 0.0001; naive versus Toca 511+5-FC). (C) Recipient athymic animals received splenocytes from either cured or naive animals and were inoculated subcutaneously with parental CT26 cells and tumor growth was monitored over time (n = 6/group) (*p = 0.04). *Statistical significance was defined as p 

Figure 8

Myeloid-Derived Suppressor Cells Decreased with…

Figure 8

Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Brain…

Figure 8
Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Brain Metastasis Model of mCRC BALB/c mice received Toca 511 pre-transduced CT26 cells intracranially and were treated with 5-FC (500 mg/kg, i.p., BID) or PBS. Treatments started at day 10 post cell inoculation and were 7 days on and 7 days off for one cycle. (A and B) The tumor (A) (*p + Gr-1+ Ly6C+ cells in the tumor (A) and spleen (B). Error bars represent SEM.
All figures (8)
Figure 8
Figure 8
Myeloid-Derived Suppressor Cells Decreased with Toca 511 and 5-FC Treatment in a Brain Metastasis Model of mCRC BALB/c mice received Toca 511 pre-transduced CT26 cells intracranially and were treated with 5-FC (500 mg/kg, i.p., BID) or PBS. Treatments started at day 10 post cell inoculation and were 7 days on and 7 days off for one cycle. (A and B) The tumor (A) (*p + Gr-1+ Ly6C+ cells in the tumor (A) and spleen (B). Error bars represent SEM.

References

    1. Ostertag D., Amundson K.K., Lopez Espinoza F., Martin B., Buckley T., Galvão da Silva A.P., Lin A.H., Valenta D.T., Perez O.D., Ibañez C.E. Brain tumor eradication and prolonged survival from intratumoral conversion of 5-fluorocytosine to 5-fluorouracil using a nonlytic retroviral replicating vector. Neuro-oncol. 2012;14:145–159.
    1. Raval R.R., Sharabi A.B., Walker A.J., Drake C.G., Sharma P. Tumor immunology and cancer immunotherapy: summary of the 2013 SITC primer. J. Immunother. Cancer. 2014;2:14.
    1. Huang T.T., Hlavaty J., Ostertag D., Espinoza F.L., Martin B., Petznek H., Rodriguez-Aguirre M., Ibañez C.E., Kasahara N., Gunzburg W. Toca 511 gene transfer and 5-fluorocytosine in combination with temozolomide demonstrates synergistic therapeutic efficacy in a temozolomide-sensitive glioblastoma model. Cancer Gene Ther. 2013;20:544–551.
    1. Huang T.T., Parab S., Burnett R., Diago O., Ostertag D., Hofman F.M., Espinoza F.L., Martin B., Ibañez C.E., Kasahara N. Intravenous administration of retroviral replicating vector, Toca 511, demonstrates therapeutic efficacy in orthotopic immune-competent mouse glioma model. Hum. Gene Ther. 2015;26:82–93.
    1. Yagiz K., Huang T.T., Lopez Espinoza F., Mendoza D., Ibañez C.E., Gruber H.E., Jolly D.J., Robbins J.M. Toca 511 plus 5-fluorocytosine in combination with lomustine shows chemotoxic and immunotherapeutic activity with no additive toxicity in rodent glioblastoma models. Neuro-oncol. 2016;18:1390–1401.
    1. Hiraoka K., Kimura T., Logg C.R., Kasahara N. Tumor-selective gene expression in a hepatic metastasis model after locoregional delivery of a replication-competent retrovirus vector. Clin. Cancer Res. 2006;12:7108–7116.
    1. Hocking C.M., Price T.J. Panitumumab in the management of patients with KRAS wild-type metastatic colorectal cancer. Therap. Adv. Gastroenterol. 2014;7:20–37.
    1. Rahmathulla G., Toms S.A., Weil R.J. The molecular biology of brain metastasis. J. Oncol. 2012;2012:723541.
    1. Norden A.D., Wen P.Y., Kesari S. Brain metastases. Curr. Opin. Neurol. 2005;18:654–661.
    1. Cohen E.E., Kane M.A., List M.A., Brockstein B.E., Mehrotra B., Huo D., Mauer A.M., Pierce C., Dekker A., Vokes E.E. Phase II trial of gefitinib 250 mg daily in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck. Clin. Cancer Res. 2005;11:8418–8424.
    1. Mongan J.P., Fadul C.E., Cole B.F., Zaki B.I., Suriawinata A.A., Ripple G.H., Tosteson T.D., Pipas J.M. Brain metastases from colorectal cancer: risk factors, incidence, and the possible role of chemokines. Clin. Colorectal Cancer. 2009;8:100–105.
    1. Mege D., Ouaissi M., Fuks D., Metellus P., Peltier J., Dufour H., Regimbeau J.M., Dahan L., Sielezneff I., Sastre B. Patients with brain metastases from colorectal cancer are not condemned. Anticancer Res. 2013;33:5645–5648.
    1. Kye B.H., Kim H.J., Kang W.K., Cho H.M., Hong Y.K., Oh S.T. Brain metastases from colorectal cancer: the role of surgical resection in selected patients. Colorectal Dis. 2012;14:e378–e385.
    1. Le D.T., Uram J.N., Wang H., Bartlett B.R., Kemberling H., Eyring A.D., Skora A.D., Luber B.S., Azad N.S., Laheru D. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015;372:2509–2520.
    1. Gabrilovich D.I., Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009;9:162–174.
    1. Ilkovitch D., Lopez D.M. The liver is a site for tumor-induced myeloid-derived suppressor cell accumulation and immunosuppression. Cancer Res. 2009;69:5514–5521.
    1. Mundy-Bosse B.L., Young G.S., Bauer T., Binkley E., Bloomston M., Bill M.A., Bekaii-Saab T., Carson W.E., 3rd, Lesinski G.B. Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4+ T cells from patients with GI malignancy. Cancer Immunol. Immunother. 2011;60:1269–1279.
    1. Wesolowski R., Markowitz J., Carson W.E., 3rd Myeloid derived suppressor cells - a new therapeutic target in the treatment of cancer. J. Immunother. Cancer. 2013;1:10.
    1. Baniyash M. Myeloid-derived suppressor cells as intruders and targets: clinical implications in cancer therapy. Cancer Immunol. Immunother. 2016;65:857–867.
    1. Vincent J., Mignot G., Chalmin F., Ladoire S., Bruchard M., Chevriaux A., Martin F., Apetoh L., Rébé C., Ghiringhelli F. 5-Fluorouracil selectively kills tumor-associated myeloid-derived suppressor cells resulting in enhanced T cell-dependent antitumor immunity. Cancer Res. 2010;70:3052–3061.
    1. Peters G.J., van der Wilt C.L. Thymidylate synthase as a target in cancer chemotherapy. Biochem. Soc. Trans. 1995;23:884–888.
    1. Cloughesy T.F., Landolfi J., Hogan D.J., Bloomfield S., Carter B., Chen C.C., Elder J.B., Kalkanis S.N., Kesari S., Lai A. Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci. Transl. Med. 2016;8:341ra75.
    1. Haraldsdottir S., Einarsdottir H.M., Smaradottir A., Gunnlaugsson A., Halfdanarson T.R. [Colorectal cancer - review] Laeknabladid. 2014;100:75–82.
    1. Zhu P., Zhao N., Sheng D., Hou J., Hao C., Yang X., Zhu B., Zhang S., Han Z., Wei L. Inhibition of growth and metastasis of colon cancer by delivering 5-fluorouracil-loaded pluronic P85 copolymer micelles. Sci. Rep. 2016;6:20896.
    1. Zang Y.W., Gu X.D., Xiang J.B., Chen Z.Y. Brain metastases from colorectal cancer: microenvironment and molecular mechanisms. Int. J. Mol. Sci. 2012;13:15784–15800.
    1. Campos S., Davey P., Hird A., Pressnail B., Bilbao J., Aviv R.I., Symons S., Pirouzmand F., Sinclair E., Culleton S. Brain metastasis from an unknown primary, or primary brain tumour? A diagnostic dilemma. Curr. Oncol. 2009;16:62–66.
    1. Noura S., Ohue M., Shingai T., Fujiwara A., Imada S., Sueda T., Yamada T., Fujiwara Y., Ohigashi H., Yano M. Brain metastasis from colorectal cancer: prognostic factors and survival. J. Surg. Oncol. 2012;106:144–148.
    1. Schouten L.J., Rutten J., Huveneers H.A., Twijnstra A. Incidence of brain metastases in a cohort of patients with carcinoma of the breast, colon, kidney, and lung and melanoma. Cancer. 2002;94:2698–2705.
    1. Yagiz K., Mendoza D., Rodriguez-Aguirre M., Espinoza F.L., Ibanez C.E., Gruber H.E., Jolly D.J., Robbins J.M. Cytotoxic and immunotherapeutic effects of Toca 511 and 5-fluorocytosine in an intraperitoneal model of metastatic colorectal cancer. Mol. Ther. 2016;24(Suppl 1):S80.
    1. Gamelin E., Delva R., Jacob J., Merrouche Y., Raoul J.L., Pezet D., Dorval E., Piot G., Morel A., Boisdron-Celle M. Individual fluorouracil dose adjustment based on pharmacokinetic follow-up compared with conventional dosage: results of a multicenter randomized trial of patients with metastatic colorectal cancer. J. Clin. Oncol. 2008;26:2099–2105.
    1. Kline C.L., Schiccitano A., Zhu J., Beachler C., Sheikh H., Harvey H.A., Mackley H.B., McKenna K., Staveley-O’Carroll K., Poritz L. Personalized dosing via pharmacokinetic monitoring of 5-fluorouracil might reduce toxicity in early- or late-stage colorectal cancer patients treated with infusional 5-fluorouracil-based chemotherapy regimens. Clin. Colorectal Cancer. 2014;13:119–126.
    1. Saif M.W., Choma A., Salamone S.J., Chu E. Pharmacokinetically guided dose adjustment of 5-fluorouracil: a rational approach to improving therapeutic outcomes. J. Natl. Cancer Inst. 2009;101:1543–1552.
    1. Vermes A., Guchelaar H.J., Dankert J. Flucytosine: a review of its pharmacology, clinical indications, pharmacokinetics, toxicity and drug interactions. J. Antimicrob. Chemother. 2000;46:171–179.
    1. Yagiz K., Rodriguez-Aguirre M.E., Espinoza F.L., Martin B., Huang T.T., Ibanez C., Ostertag D., Kasahara N., Gruber H.E., Jolly D.J. Intravenous delivery of Toca 511 gene therapy in combination with 5-fluorocytosine for intratumoral production of 5-fluorouracil in a colon cancer metastasis model. Mol. Ther. 2015;23(Suppl 1):S213.
    1. Kanterman J., Sade-Feldman M., Biton M., Ish-Shalom E., Lasry A., Goldshtein A., Hubert A., Baniyash M. Adverse immunoregulatory effects of 5FU and CPT11 chemotherapy on myeloid-derived suppressor cells and colorectal cancer outcomes. Cancer Res. 2014;74:6022–6035.
    1. Otvos B., Silver D.J., Mulkearns-Hubert E.E., Alvarado A.G., Turaga S.M., Sorensen M.D., Rayman P., Flavahan W.A., Hale J.S., Stoltz K. Cancer stem cell-secreted macrophage migration inhibitory factor stimulates myeloid derived suppressor cell function and facilitates glioblastoma immune evasion. Stem Cells. 2016;34:2026–2039.
    1. Marigo I., Dolcetti L., Serafini P., Zanovello P., Bronte V. Tumor-induced tolerance and immune suppression by myeloid derived suppressor cells. Immunol. Rev. 2008;222:162–179.
    1. Suzuki E., Kapoor V., Jassar A.S., Kaiser L.R., Albelda S.M. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin. Cancer Res. 2005;11:6713–6721.
    1. Ko H.J., Kim Y.J., Kim Y.S., Chang W.S., Ko S.Y., Chang S.Y., Sakaguchi S., Kang C.Y. A combination of chemoimmunotherapies can efficiently break self-tolerance and induce antitumor immunity in a tolerogenic murine tumor model. Cancer Res. 2007;67:7477–7486.
    1. Mitchell L.A., Lopez Espinoza F., Mendoza D., Kato Y., Inagaki A., Hiraoka K., Kasahara N., Gruber H.E., Jolly D.J., Robbins J.M. Toca 511 gene transfer and treatment with the prodrug, 5-fluorocytosine, promotes durable antitumor immunity in a mouse glioma model. Neuro-oncol. 2017;19:930–939.
    1. Monu N.R., Frey A.B. Myeloid-derived suppressor cells and anti-tumor T cells: a complex relationship. Immunol. Invest. 2012;41:595–613.
    1. Raychaudhuri B., Rayman P., Huang P., Grabowski M., Hambardzumyan D., Finke J.H., Vogelbaum M.A. Myeloid derived suppressor cell infiltration of murine and human gliomas is associated with reduction of tumor infiltrating lymphocytes. J. Neurooncol. 2015;122:293–301.
    1. Ribechini E., Greifenberg V., Sandwick S., Lutz M.B. Subsets, expansion and activation of myeloid-derived suppressor cells. Med. Microbiol. Immunol. (Berl.) 2010;199:273–281.
    1. Raber P.L., Thevenot P., Sierra R., Wyczechowska D., Halle D., Ramirez M.E., Ochoa A.C., Fletcher M., Velasco C., Wilk A. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int. J. Cancer. 2014;134:2853–2864.
    1. Movahedi K., Guilliams M., Van den Bossche J., Van den Bergh R., Gysemans C., Beschin A., De Baetselier P., Van Ginderachter J.A. Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood. 2008;111:4233–4244.
    1. Youn J.I., Nagaraj S., Collazo M., Gabrilovich D.I. Subsets of myeloid-derived suppressor cells in tumor-bearing mice. J. Immunol. 2008;181:5791–5802.
    1. Parker K.H., Beury D.W., Ostrand-Rosenberg S. Myeloid-derived suppressor cells: critical cells driving immune suppression in the tumor microenvironment. Adv. Cancer Res. 2015;128:95–139.
    1. Umansky V., Sevko A. Tumor microenvironment and myeloid-derived suppressor cells. Cancer Microenviron. 2013;6:169–177.
    1. Meyerhardt J.A., Mayer R.J. Systemic therapy for colorectal cancer. N. Engl. J. Med. 2005;352:476–487.
    1. McCormack P.L., Keam S.J. Bevacizumab: a review of its use in metastatic colorectal cancer. Drugs. 2008;68:487–506.
    1. Razenberg L.G., van Gestel Y.R., de Hingh I.H., Loosveld O.J.L., Vreugdenhil G., Beerepoot L.V., Creemers G.J., Lemmens V.E. Bevacizumab for metachronous metastatic colorectal cancer: a reflection of community based practice. BMC Cancer. 2016;16:110.
    1. Grigorean V.T., Ciuhu A.N., Rahnea Nita G., Strambu V., Straja D.N., Popescu M., Sandu A.M., Rahnea Nita R.A. Efficacy of cetuximab in metastatic colon cancer - case report. Chirurgia (Bucur.) 2014;109:383–389.
    1. Price T.J., Peeters M., Kim T.W., Li J., Cascinu S., Ruff P., Suresh A.S., Thomas A., Tjulandin S., Zhang K. Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): a randomised, multicentre, open-label, non-inferiority phase 3 study. Lancet Oncol. 2014;15:569–579.
    1. Foubert F., Matysiak-Budnik T., Touchefeu Y. Options for metastatic colorectal cancer beyond the second line of treatment. Dig. Liver Dis. 2014;46:105–112.
    1. Perez O.D., Logg C.R., Hiraoka K., Diago O., Burnett R., Inagaki A., Jolson D., Amundson K., Buckley T., Lohse D. Design and selection of Toca 511 for clinical use: modified retroviral replicating vector with improved stability and gene expression. Mol. Ther. 2012;20:1689–1698.

Source: PubMed

3
Subskrybuj