Journal List > Immune Netw > v.16(2) > 1033538

Kim, Kim, Park, Kim, Lee, Hong, Kim, and Han: Cell-based Immunotherapy for Colorectal Cancer with Cytokine-induced Killer Cells

Abstract

Colorectal cancer is the third leading cancer worldwide. Although incidence and mortality of colorectal cancer are gradually decreasing in the US, patients with metastatic colorectal cancer have poor prognosis with an estimated 5-year survival rate of less than 10%. Over the past decade, advances in combination chemotherapy regimens for colorectal cancer have led to significant improvement in progression-free and overall survival. However, patients with metastatic disease gain little clinical benefit from conventional therapy, which is associated with grade 3~4 toxicity with negative effects on quality of life. In previous clinical studies, cell-based immunotherapy using dendritic cell vaccines and sentinel lymph node T cell therapy showed promising therapeutic results for metastatic colorectal cancer. In our preclinical and previous clinical studies, cytokine-induced killer (CIK) cells treatment for colorectal cancer showed favorable responses without toxicities. Here, we review current treatment options for colorectal cancer and summarize available clinical studies utilizing cell-based immunotherapy. Based on these studies, we recommend the use CIK cell therapy as a promising therapeutic strategy for patients with metastatic colorectal cancer.

Abbreviations

CIK cells

cytokine-induced killer cells

CRC

colorectal cancer

LOH

loss of heterozygosity

CIN

chromosomal instability

MSI

microsatellite instability

MMR

mismatch repair

DFS

disease-free survival

OS

overall survival

5-FU

5-fluorouracil

LV

leucovorin

EGFR

epidermal growth factor receptor

VEGF

vascular endothelial growth factor

DCs

dendritic cells

CEA

carcinoembryonic antigen

MAGE

melanoma-associated antigen

ACT

adoptive cell therapy

TILs

tumor-infiltrating lymphocytes

SLN

sentinel lymph node

INTRODUCTION

Colorectal cancer (CRC) is one of the most prevalent cancer, along with lung and breast cancers. In 2012, approximately 1.4 million new cases and 700,000 CRC-related deaths were estimated worldwide (1). Over the last decade, CRC incidence and mortality in the United States have been on a steady decline as a result of advances in its prevention, screening, and treatment. Nevertheless, it is still the second leading cause of cancer deaths (2). Moreover, there has been a significant rise in CRC incidence in newly developed or economically transitioning countries, such as South America, Eastern European countries, and most parts of Asia (3). Although CRC is highly curable at an early stage, approximately 50% of patients with CRCs eventually develop metastatic disease (4). Five-year survival rate is approximately 10% in patients with a stage IV disease (5).
There are many known risk factors for CRC including age, sex, smoking, excessive alcohol consumption, high intake of red and processed meat, obesity, diabetes, inflammatory bowel disease, inherited genetic disorder, and family history of CRC (6789101112). Among them, the most significant risk factor is age. Incidence significantly increases in adults 50 years and older (13). Furthermore, inflammatory bowel disease—ulcerative colitis and Crohn's disease—increase the overall risk of colitis-associated cancer (14). A link between inflammation and cancer is well-established. Approximately 20% of patients with inflammatory bowel disease develop colitis-associated cancer within 30 years of disease onset (15). Inherited CRC, including Lynch syndrome and familial adenomatous polyposis, represent less than 5% of CRC cases (16).
In this review, we briefly review currently available drugs for the treatment of colorectal cancer and summarize clinical studies that use cell-based cancer immunotherapy as promising therapeutic strategy. We also suggest the use of cytokine-induced killer (CIK) cells as an additional therapeutic approach for metastatic colorectal cancer.

THE MOLECULAR PATHOGENESIS OF COLORECTAL CANCER

Colorectal cancer is a heterogeneous disease. Most CRCs occur without a known genetic risk factor and significant family history. Only a small proportion (5%) of inherited CRCs, which is also known as familial CRC, have been thoroughly characterized (17), and additional 20~25% are yet to be completely understood (17). CRC develops over the course of more than 10 years, and entails multistep genetic events for tumor progression. A Multistep genetic model of colorectal carcinogenesis was proposed by Fearon and Vogelstein. Colorectal cancers occur as a result of oncogenes activation, along with the inactivation of tumor suppressor genes by mutations or loss of heterozygosity (LOH) (18). Mutations in at least four to five genes are required for the formation of a malignant tumor (18).
The molecular pathogenesis of colorectal cancer is commonly classified into two main pathways of genomic instability: chromosomal instability (CIN) and microsatellite instability (MSI) pathways. CIN leads to an imbalance in chromosome number (aneuploidy) such as chromosome gains or losses (including LOH events at specific tumor suppressor gene loci), and is observed in 80~85% of sporadic CRCs (18192021). A mutation coupled with LOH of the adenomatous polyposis coli (APC) tumor suppressor gene, followed by activating mutations of v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS), lead to the initial appearance of adenoma from normal colonic epithelium (18). APC mutations are typically observed in earliest stages of tumors, including in adenomas as small as 0.5 cm (22). This indicates that a functional loss of APC, which is part of the Wnt signaling pathway, plays a role in early tumorigenesis. In contrast, MSI, which is observed in 15~20% of sporadic CRCs (21), is a hypermutable phenotype caused by impaired DNA mismatch repair (MMR) system to correct errors such as single-base mismatches and insertion-deletion loops that spontaneously occur during DNA replication (2324). MSI arises from impaired DNA MMR genes, such as MutL homolog 1 (MLH) 1, MutS homolog (MSH) 2, and to a lesser degree MSH6 (2526). Faulty DNA fidelity caused by defects in DNA MMR causes frameshift and point mutations, mainly in repetitive sequences (microsatellites) (26). Lynch syndrome is a well-established hereditary predisposition to colorectal cancer caused by a germline mutation in a DNA MMR gene. The syndrome accounts for approximately 3% of all CRCs (16). However, 12% of MSI exhibit sporadic CRC with CIN (27). Because pathways are not mutually exclusive, CRC can display features of multiple pathways. The significance of these features is not fully understood (28).

CURRENT TREATMENT OF COLORECTAL CANCER

Surgery is the cornerstone treatment for colorectal cancer. In stage I colon cancer, surgery is the definitive treatment without the use of adjuvant chemotherapy. Benefit of adjuvant chemotherapy for patients with a stage II disease is highly debatable due to minimal gains in overall response. However, adjuvant chemotherapy is generally acceptable for the use patients with node-positive cancer following surgery (high-risk stage II disease) (29). Adjuvant chemotherapy is required for all patients with a stage III colon cancer after surgical resection, which have a high risk of recurrence of 15~50% (30). Conventional chemotherapeutic agents include 5-fluorouracil (5-FU) with leucovorin (LV), capecitabine (an orally administered prodrug of 5-FU), oxaliplatin, and irinotecan. Adjuvant chemotherapy using the 5-FU/LV regimens following surgery provides significant reduction in mortality by 22% and improves event-free survival by 35% (31). To improve disease-free survival (DFS) and overall survival (OS), currently, FOLFOX (5-FU/LV and oxaliplatin) or FOLFIRI (5-FU/LV and irinotecan) is widely used for standard first- or second-line treatment in patients with node-positive colon cancer following surgical resection (3233). However, FOLFOX and FOLFIRI demonstrated greater toxicities, including grade 3 or higher acute toxicity, compared to 5-FU/LV alone (34).
In 2004, targeted therapeutic agents for metastatic colorectal cancer, such as anti-epidermal growth factor receptor (EGFR) antibodies (cetuximab and panitumumab), vascular endothelial growth factor (VEGF) inhibitors (bevacizumab, ziv-aflibercept, and ramucirumab), and a multikinase inhibitor (regorafenib), were approved by the US Food and Drug Administration (FDA) for combinational use with standard chemotherapy treatment (35). Currently, there are various regimens of combinational therapies using targeted therapeutic agents and conventional chemotherapy drugs (36). Treatment strategy is often dependent on tumor-specific factors, such as KRAS and/or v-Raf murine sarcoma viral oncogene homolog B (BRAF) mutation, and condition of patients and their disease progression following first-line treatment (3336). Since RAS mutations lead to continuous activation of signaling pathways downstream of EGFR, the use of anti-EGFR antibodies is effective in patients with wild-type KRAS exon 2, but not in patients with KRAS and/or BRAF mutations. Thus, approximately 30~40% of CRCs are associated with KRAS gene mutation and do not respond to anti-EGFR antibodies (37). Over the past decade, multidisciplinary advances in the treatment of metastatic colorectal cancer with targeted therapy have statistically improved progression-free and overall survivals. Despite these advances, only a relatively small effect on survival outcomes was observed, especially in patients with distant metastatic disease. Therefore, development of new therapeutic strategy against metastatic colorectal cancer is urgently needed.

DENDRITIC CELL THERAPY FOR COLORECTAL CANCER

Dendritic cell (DC) therapy is one of several strategies in therapeutic cancer vaccines. Sipuleucel-T, a DC-based cancer vaccine, has been approved by the US FDA for advanced prostate cancer in 2011. The goal of DC vaccination is to elicit anti-tumor response by inducing tumor-specific effector T cells. DCs are generated ex vivo by culturing hematopoietic progenitor cells or monocytes with a combination of cytokines and then pulsing them with antigens ex vivo (38). Tumor antigen choice for DCs loading is crucial to achieve optimal clinical outcomes. There are various type of cancer antigens for DCs loading, including mutated and non-mutated antigens (39).
Now, we can find 9 clinical trials for the treatment of patients with CRC from 2004 to 2015 (Table 1). Antigens that are most commonly used for colorectal cancer include carcinoembryonic antigen (CEA) peptides (4041), melanoma-associated antigen (MAGE) from allogeneic melanoma cell lysates (4243), and autologous cell lysates from biopsy material (44). While CEA expression in normal colon epithelial cells is relatively low, it is over expressed in most colorectal carcinomas as well as in many cancers (45). Therefore, CEA has been the major immunological target of DC-based cancer vaccines for colorectal cancer. Clinical trials of CEA-pulsed DCs demonstrated its immune-stimulatory capacity and it was well tolerated in patients without any observable toxicities. However, the overall clinical response was rather unimpressive (404146), which may reflect severely impaired immune functions in patients with excessive tumor burdens and tumor immunoediting mechanisms. Nevertheless, several clinical studies of MAGE-pulsed DCs showed 24~40% clinical benefit rate with durable responses and tumor regression (4243). Moreover, regulatory T cell levels declined upon DC vaccination (42). Recently, Hunyadi J. and colleagues demonstrated that autologous tumor cell lysates-loaded DCs led to an increase in 6-years survival rate in colorectal cancer patients and more efficient induction of T-lymphocytes proliferation in vitro when compared to CEA-pulsed DCs (44). However, because of limited number of patients, additional evaluations in large-scale clinical trials are needed. Although significant advances have been made over the past decade, further studies are required to fully determine the potential antitumor effects of DC vaccination for colorectal cancer.

T CELL THERAPY OF COLORECTAL CANCER

Adoptive cell therapy (ACT) for metastatic melanoma was first described in 1988 (47). In ACT, tumor-infiltrating lymphocytes (TILs) are collected from solid tumor specimen, and are activated and expanded ex vivo. Subsequently, TILs are administered intravenously to the autologous patient. In 2002, Dudley ME et al. demonstrated that host immunodepletion using cyclophosphamide and fludarabine prior to TIL infusion resulted in dramatic improvement in clinical outcomes for patients with metastatic melanoma (48). Although ACT has been evaluated in various cancers, only a small number of ACT trials for CRC have been performed. In a phase I clinical trial, 14 patients with colorectal liver metastases received ACT with TILs in combination with high-dose IL-2 (49). Unfortunately, no significant difference in DFS rates was observed between TILs therapy and conventional chemotherapy. A novel ACT approach for CRC was conducted in two separate studies; sentinel lymph node (SLN) T lymphocytes were used in a pilot study (seven with stage II-III and nine with stage IV CRC) (50) and in a phase I/II study (46 with stage I-III and nine with stage IV CRC) (51), respectively. The pilot study proposed feasible clinical outcomes in patients with CRC (50). All patients with stage IV disease responded to the treatment with the following outcomes: four stable disease, one partial response, and four complete response with no detectable cancer cells remaining. The overall survival of ACT-treated patients was also significantly improved to 2.6 years compared to 0.8 years in conventionally-treated control patients. The latest phase I/II study also showed favorable clinical outcomes (51). The median overall survival of SLN-T lymphocytes-treated and control groups in stage IV patients were 28 and 14 months, respectively. In the two studies, no significant toxicity was observed following SLN-T lymphocytes treatment. These studies demonstrated that SLN-T lymphocyte immunotherapy is indeed feasible and safe for patients with metastatic CRC. However, surgeon's proficiency may be required for the intraoperative sentinel node detection. In a phase I study of autologous genetically engineered T cells, all patients developed severe colitis despite showing a decrease in serum CEA levels by CEA-specific T cells (52).

CYTOKINE-INDUCED KILL CELL THERAPY FOR COLORECTAL CANCER

Cytokine-induced killer (CIK) cells are ex vivo-expanded lymphocytes used for cancer immunotherapy. CIK cells are generated by culturing peripheral blood mononuclear cells (PBMCs) with a combination of IL-2 and anti-CD3 monoclonal antibodies for 14 days (53). Subsequently, CIK cells are harvested and administered intravenously to the autologous patient. CIK cells consist of heterogeneous cell population, mainly CD3+CD56, CD3+CD56+, and a minor population of CD3CD56+ cells. Importantly, CD3+CD56+ cells are the distinguishing cell population among the CIK cells and have the most potent cytotoxic function (53). CD3+CD56+ cells originated from CD3+CD8+CD56 cells but not from CD3+CD56+ cells during the ex vivo expansion (54). CIK cell cytotoxicity is mediated by perforin release and dependent on several activating receptors such as NKG2D, NKp30, and DNAM-1 (5556). CIK cells also exhibit non-specific and non-MHC-restricted cytotoxicity (56).
Over the past decade, CIK cell therapy has been evaluated in numerous clinical studies in patients with various types of cancer, such as hepatocellular carcinoma, non-small cell lung cancer, renal cell carcinoma, and gastric cancer (57). CIK cell therapy can be used as a postoperative adjuvant treatment as well as a palliative treatment following standard therapies. CIK cell therapy was evaluated in a limited number of clinical studies in patients with colorectal cancer. In a retrospective study, 21 of 96 colorectal cancer patients who underwent surgery as well as adjuvant chemotherapy received one to three cycles of CIK cell transfusion for immunotherapy (58). Patients in the CIK-treated group had significant improvement in their 2-year DFS rates than those in the control group (59.65±24.80% vs 29.35±6.39%). CIK cell transfusions were well tolerated without any observable toxicity. Other studies used CIK in a combination therapy with DCs, specifically tumor lysate-pulsed DCs. In 2014, two clinical studies of DC vaccine and CIK cell combinational therapy for colorectal cancer patients were published. One study demonstrated that overall survival rates were significantly improved in the DC-CIK therapy compared to the control group (p=0.04) (59). Moreover, most patients that received DC-CIK therapy displayed improvement in quality of life, including physical strength, appetite, sleep, and body weight. The adverse effects were mild and self-resolving. In the other study, Gao, D. and colleagues evaluated the clinical benefits of DC-CIK therapy in 54 gastric and colorectal patients following surgery with or without chemo-radiotherapy (60). The study demonstrated that DFS and 5-year survival of colorectal cancer patients were significantly prolonged in DC-CIK treatment groups (DFS rate: 66% and 5-year survival rates: 75%) when compared to patients in control groups (DFS rate: 8% and 5-year survival rates: 15%; p<0.01). Serum levels of cytokines were also evaluated. IFN-γ and IL-12 levels were significantly increased in patients that received the DC-CIK treatment. Besides fever, no adverse events were observed in patients that received the DC-CIK therapy. Since they did not compare the efficacy of combination therapy with each alone, we could not clarify whether combination therapy is better than each therapy. However, when we considered the efficacy of the single therapy of DCs and CIK cells described above, we could presume that The combination of CIK and DC might exert better antitumor activity than each alone.
Here, we provide additional evidence that CIK cell therapy can effectively prevent growth of colorectal cancers in a xenograft mouse model. CIK cells were generated from PBMCs of healthy volunteers. PBMCs were isolated by Ficoll–Hypaque density gradient centrifugation, washed three times with PBS, and cultured in the presence of immobilized anti-CD3 antibody (5 µg/ml) and recombinant human IL-2 (700 U/ml) for five days. The cell suspension was further incubated in complete medium containing IL-2 only (170 U/ml) for nine days. Thereafter, medium containing IL-2 was replenished every 2 or 3 days. On day 14, cell phenotypes were examined by flow cytometry. CIK cells comprised of 94% CD3+, 3% CD3CD56+, 16% CD3+CD56+, 14% CD4+, and 81% CD8+ cells (Fig. 1A). To examine the cytotoxicity of CIK cells, two cell lines, SW620 and K-562 were used. SW620 cells are metastatic human colon cancer cell lines derived from lymph node (61). K-562 cell lines are human leukemic cells commonly used as reference target cells of NK cells due to the very low level of MHC class I antigens on their cell membrane (62). A 4-h 51Cr release assay revealed that CIK cells were able to kill 14%, 22%, and 36% of SW620 cells at effector:target ratios of 10:1, 30:1, and 100:1, respectively (Fig. 1B). Moreover, CIK cells showed strong cytotoxicity against K562 cells, a known NK-sensitive target.
A nude mouse xenograft assay was used to examine the in vivo antitumor activity of CIK cells. SW620 cells (6×106) in 300 µl of PBS were injected subcutaneously on day 0, followed by intravenous once-a-week injection of CIK cells and adriamycin (ADR). On day 25, mice were sacrificed and the tumor mass and body weight (to determine toxicity) were measured. In control mice, SW620 cells grew to a tumor volume of 473±190 mm3 (n=7) (Fig. 1C). A strong dose-dependent anti-tumor effect of CIK cells was observed. CIK cells injected at doses of 1×106, 3×106, and 10×106 cells per mouse were able to inhibit tumor growth by 7%, 53%, and 73%, respectively. ADR also strongly inhibited the growth of SW620 cells (Fig. 1C). In control mice, weight of SW620 cells reached 1,676±530 mg at 25 days post-implantation. CIK cells did not affect body weight gain of nude mice (Fig. 1D). CIK cells injected at doses of 1×106, 3×106 and 10×106 cells per mouse reduced tumor weight by 10%, 54% and 73%, respectively (Fig. 1E). Thus, our preclinical efficacy data showed that CIK cells were able to kill SW620 cells in vitro and in vivo, suggesting that CIK cells might be a good immunotherapy candidate for colorectal cancer.

CONCLUSION

Patients with metastatic colorectal cancer have significant risk of recurrence following surgery and conventional adjuvant therapy. Although advances in therapeutic agents for CRC have reduced the risk of recurrence and increased overall survival, patients with metastatic disease have a poor prognosis and a particularly low 5-year survival rates of less than 10%. Additionally, current combination therapy is occasionally discontinued due to a grade 3~4 toxicity with negative effects on quality of life (63). Overall quality of life should be considered for patients with stage IV disease for palliative therapy. Also, elderly patients with stage III disease often avoid adjuvant chemotherapy due to anxiety of old age, comorbidities, side effects, and perceived minimal benefit (64).
Cell-based cancer immunotherapy is a promising therapeutic strategy. Clinical and preclinical studies of CRC treatment with DCs, T cells, and CIK cells showed promising outcomes, although only limited information have been available till recently. Among the immune effector cells, CIK cells have certain advantages in a clinical application: they are relatively easy to generate and expanded in large-scale from PBMCs, their non-MHC-restricted cytotoxic activity could eliminate MHC class I-negative tumor cells, and importantly, CIK cell therapy causes mild, transient, and easy-to-manage side effects. Over the last decades, CIK cell therapy has been evaluated in numerous clinical studies in patients with various cancers. In most patients, combination therapy using conventional agents and CIK cells showed superior clinical outcomes than standard therapy alone. Clinical studies previously reported by others and our preclinical data suggest that immunotherapy of CRC with CIK cells can be a promising strategy to limit the growth and metastasis of CRC.

Figures and Tables

Figure 1

Antitumor efficacy of cytokine-induced killer (CIK) cells for CRC in a mouse xenograft model. CIK cells were generated by culturing human peripheral blood mononuclear cells (PBMCs) with a combination of IL-2 and anti-CD3 antibody for 14 days. CIK cells were stained with human monoclonal antibodies against CD3, CD56, CD4, and CD8 (A). In vitro cytotoxicity of CIK cells was evaluated by a 4-h 51Cr-release assay that used SW620 and K562 as target cells (B). Effector and target cells (1×104 cells/100 µl/well) were mixed at different ratios (1:1 to 100:1). The percentage of cytotoxicity was calculated as following: cytotoxicity = [(sample-spontaneous)/(maximum-spontaneous)]×100. Spontaneous release was measured from target cells incubated in medium alone, whereas maximum release was measured after cells were treated with 2% Nonidet P-40. Nude mice (n=7) were implanted subcutaneously with 6×106 SW620 cells. CIK cells (1 to 10×106 cells/mouse) were injected intravenously once a week. Adriamycin (ADR) was injected intravenously at 2 mg/kg. Tumor size was estimated as length (mm)×width (mm)×height (mm)/2 (C). Body weight was measured to estimate toxicity (D). Mice were sacrificed on day 25 and tumors were weighed (E). Statistical significance was determined by Student's t-test versus the PBS-treated control group (*p<0.05, **p<0.01). All experimental procedures were approved by the Animal Experimentation Ethics Committee and by the Institutional Ethics Committee of Chungbuk National University. Informed consents have been obtained from volunteers.

in-16-99-g001
Table I

Summary of clinical studies of dendritic cell-based immunotherapy for CRC (a search of the PubMed from 2004 to 2015)

in-16-99-i001
Author Cell types Number of patients Clinical outcome Adverse events
Hunyadi et al. (44) DC+autologous tumor cell lysates 6 patients Survival for 6-years (5 of 6) No adverse effects
DC+CEA peptide 6 patients Survival for 6-years (3 of 3)
Morse et al. (65) DC+Poxvectors encoding CEA and MUC1 (PANVAC) 37 patients 2 of 37 death No grade 3/4 toxicity except grade 3 urticaria on the DC+PANVAC
PANVAC+GM-CSF 37 patients 5 of 37 death
Sakakibara et al. (41) DC+CEA peptide 10 patients PD (7), SD (1), withdrawal (2) No adverse effects except grade I fever
Toh et al. (42) DC+MAGE 20 patients PD (11), PR (1), SD (7), withdrawal (1) Mild grade 1 or 2 toxicity
Burgdorf et al. (43) DC+MAGE 20 patients 24% of SD (4) No adverse effects
Kavanagh et al. (46) DC+CEA peptide 21 patients 11 of all patients evaluated, PD (11) No significant toxicity
Morse et al. (66) DC+Fowlpox vector encoding CEA and costimulatory molecules 11 patients, 3 patients (NSCLC) PD (8), SD/MR (6) No grade 3/4 toxicity
Liu et al. (40) DC+CEA peptide 10 patients PD (8), SD (2) No grade 2/3 toxicity
Matsuda et al. (67) DC+CEA peptide 7 patients PD (5), SD (2) No adverse effects

DC, dendritic cell; CEA, carcinoembryonic antigen; GM-CSF, granulocyte-macrophage colony-stimulating factor; PD, progressive disease; SD, stable disease; PR, partial response; MR, minor response; MAGE, melanoma-associated antigen; CRC, colorectal cancer; NSCLC, non-small cell lung cancer.

Abbreviations

CIK cells

cytokine-induced killer cells

CRC

colorectal cancer

LOH

loss of heterozygosity

CIN

chromosomal instability

MSI

microsatellite instability

MMR

mismatch repair

DFS

disease-free survival

OS

overall survival

5-FU

5-fluorouracil

LV

leucovorin

EGFR

epidermal growth factor receptor

VEGF

vascular endothelial growth factor

DCs

dendritic cells

CEA

carcinoembryonic antigen

MAGE

melanoma-associated antigen

ACT

adoptive cell therapy

TILs

tumor-infiltrating lymphocytes

SLN

sentinel lymph node

ACKNOWLEDGEMENTS

This work was supported by the research grant of the Chungbuk National University in 2014.

Notes

CONFLICTS OF INTEREST The authors have no financial conflict of interest.

References

1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D, Bray F. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015; 136:E359–E386.
crossref
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015; 65:5–29.
crossref
3. Center MM, Jemal A, Ward E. International trends in colorectal cancer incidence rates. Cancer Epidemiol Biomarkers Prev. 2009; 18:1688–1694.
crossref
4. Kindler HL, Shulman KL. Metastatic colorectal cancer. Curr Treat Options Oncol. 2001; 2:459–471.
crossref
5. Sanoff HK, Sargent DJ, Campbell ME, Morton RF, Fuchs CS, Ramanathan RK, Williamson SK, Findlay BP, Pitot HC, Goldberg RM. Five-year data and prognostic factor analysis of oxaliplatin and irinotecan combinations for advanced colorectal cancer: N9741. J Clin Oncol. 2008; 26:5721–5727.
crossref
6. Cho E, Smith-Warner SA, Ritz J, van den Brandt PA, Colditz GA, Folsom AR, Freudenheim JL, Giovannucci E, Goldbohm RA, Graham S, Holmberg L, Kim DH, Malila N, Miller AB, Pietinen P, Rohan TE, Sellers TA, Speizer FE, Willett WC, Wolk A, Hunter DJ. Alcohol intake and colorectal cancer: a pooled analysis of 8 cohort studies. Ann Intern Med. 2004; 140:603–613.
7. Liang PS, Chen TY, Giovannucci E. Cigarette smoking and colorectal cancer incidence and mortality: systematic review and meta-analysis. Int J Cancer. 2009; 124:2406–2415.
crossref
8. Chan DS, Lau R, Aune D, Vieira R, Greenwood DC, Kampman E, Norat T. Red and processed meat and colorectal cancer incidence: meta-analysis of prospective studies. PLoS One. 2011; 6:e20456.
crossref
9. Ma Y, Yang Y, Wang F, Zhang P, Shi C, Zou Y, Qin H. Obesity and risk of colorectal cancer: a systematic review of prospective studies. PLoS One. 2013; 8:e53916.
crossref
10. Jiang Y, Ben Q, Shen H, Lu W, Zhang Y, Zhu J. Diabetes mellitus and incidence and mortality of colorectal cancer: a systematic review and meta-analysis of cohort studies. Eur J Epidemiol. 2011; 26:863–876.
crossref
11. Jess T, Rungoe C, Peyrin-Biroulet L. Risk of colorectal cancer in patients with ulcerative colitis: a meta-analysis of population-based cohort studies. Clin Gastroenterol Hepatol. 2012; 10:639–645.
crossref
12. Taylor DP, Burt RW, Williams MS, Haug PJ, Cannon-Albright LA. Population-based family history-specific risks for colorectal cancer: a constellation approach. Gastroenterology. 2010; 138:877–885.
crossref
13. Haggar FA, Boushey RP. Colorectal cancer epidemiology: incidence, mortality, survival, and risk factors. Clin Colon Rectal Surg. 2009; 22:191–197.
crossref
14. von Roon AC, Reese G, Teare J, Constantinides V, Darzi AW, Tekkis PP. The risk of cancer in patients with Crohn's disease. Dis Colon Rectum. 2007; 50:839–855.
crossref
15. Lakatos PL, Lakatos L. Risk for colorectal cancer in ulcerative colitis: changes, causes and management strategies. World J Gastroenterol. 2008; 14:3937–3947.
crossref
16. Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Clendenning M, Sotamaa K, Prior T, Westman JA, Panescu J, Fix D, Lockman J, LaJeunesse J, Comeras I, de la Chapelle A. Feasibility of screening for Lynch syndrome among patients with colorectal cancer. J Clin Oncol. 2008; 26:5783–5788.
crossref
17. Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology. 2010; 138:2044–2058.
crossref
18. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell. 1990; 61:759–767.
crossref
19. Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. Nature. 1998; 396:643–649.
crossref
20. Lothe RA, Peltomaki P, Meling GI, Aaltonen LA, Nystrom-Lahti M, Pylkkanen L, Heimdal K, Andersen TI, Moller P, Rognum TO, Fossa SD, Haldorsen T, Langmark F, Brogger A, de la Chapelle A, Borresen A. Genomic instability in colorectal cancer: relationship to clinicopathological variables and family history. Cancer Res. 1993; 53:5849–5852.
21. Grady WM, Carethers JM. Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterology. 2008; 135:1079–1099.
crossref
22. Powell SM, Zilz N, Beazer-Barclay Y, Bryan TM, Hamilton SR, Thibodeau SN, Vogelstein B, Kinzler KW. APC mutations occur early during colorectal tumorigenesis. Nature. 1992; 359:235–237.
crossref
23. Aaltonen LA, Peltomaki P, Leach FS, Sistonen P, Pylkkanen L, Mecklin JP, Jarvinen H, Powell SM, Jen J, Hamilton SR, Petersen GM, Kinzler KW, Vogelstein B, de la Chapellet A. Clues to the pathogenesis of familial colorectal cancer. Science. 1993; 260:812–816.
crossref
24. Ionov Y, Peinado MA, Malkhosyan S, Shibata D, Perucho M. Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature. 1993; 363:558–561.
crossref
25. Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, Jessup JM, Kolodner R. Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res. 1997; 57:808–811.
26. Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B, Starling N. Colorectal cancer. Lancet. 2010; 375:1030–1047.
crossref
27. Shen L, Toyota M, Kondo Y, Lin E, Zhang L, Guo Y, Hernandez NS, Chen X, Ahmed S, Konishi K, Hamilton SR, Issa JP. Integrated genetic and epigenetic analysis identifies three different subclasses of colon cancer. Proc Natl Acad Sci U S A. 2007; 104:18654–18659.
crossref
28. Pino MS, Chung DC. The chromosomal instability pathway in colon cancer. Gastroenterology. 2010; 138:2059–2072.
crossref
29. Benson AB 3rd, Schrag D, Somerfield MR, Cohen AM, Figueredo AT, Flynn PJ, Krzyzanowska MK, Maroun J, McAllister P, Van Cutsem E, Brouwers M, Charette M, Haller DG. American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol. 2004; 22:3408–3419.
crossref
30. Brenner H, Kloor M, Pox CP. Colorectal cancer. Lancet. 2014; 383:1490–1502.
crossref
31. Efficacy of adjuvant fluorouracil and folinic acid in colon cancer. International Multicentre Pooled Analysis of Colon Cancer Trials (IMPACT) investigators. Lancet. 1995; 345:939–944.
32. Tournigand C, Andre T, Achille E, Lledo G, Flesh M, Mery-Mignard D, Quinaux E, Couteau C, Buyse M, Ganem G, Landi B, Colin P, Louvet C, de Gramont A. FOLFIRI followed by FOLFOX6 or the reverse sequence in advanced colorectal cancer: a randomized GERCOR study. J Clin Oncol. 2004; 22:229–237.
crossref
33. Andrea C, Fausto P, Francesca BK, Mary C, Mara G, Veronica L, Sandro B. Which strategy after first-line therapy in advanced colorectal cancer? World J Gastroenterol. 2014; 20:8921–8927.
34. Braun MS, Seymour MT. Balancing the efficacy and toxicity of chemotherapy in colorectal cancer. Ther Adv Med Oncol. 2011; 3:43–52.
crossref
35. Hohla F, Winder T, Greil R, Rick FG, Block NL, Schally AV. Targeted therapy in advanced metastatic colorectal cancer: current concepts and perspectives. World J Gastroenterol. 2014; 20:6102–6112.
crossref
36. Fakih MG. Metastatic colorectal cancer: current state and future directions. J Clin Oncol. 2015; 33:1809–1824.
crossref
37. Ahmed S, Johnson K, Ahmed O, Iqbal N. Advances in the management of colorectal cancer: from biology to treatment. Int J Colorectal Dis. 2014; 29:1031–1042.
crossref
38. Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, Oh S, Fay J, Pascual V, Banchereau J, Palucka K. Harnessing human dendritic cell subsets for medicine. Immunol Rev. 2010; 234:199–212.
crossref
39. Obeid J, Hu Y, Slingluff CL Jr. Vaccines, adjuvants, and dendritic cell activators--current status and future challenges. Semin Oncol. 2015; 42:549–561.
crossref
40. Liu KJ, Wang CC, Chen LT, Cheng AL, Lin DT, Wu YC, Yu WL, Hung YM, Yang HY, Juang SH, Whang-Peng J. Generation of carcinoembryonic antigen (CEA)-specific T-cell responses in HLA-A*0201 and HLA-A*2402 late-stage colorectal cancer patients after vaccination with dendritic cells loaded with CEA peptides. Clin Cancer Res. 2004; 10:2645–2651.
crossref
41. Sakakibara M, Kanto T, Hayakawa M, Kuroda S, Miyatake H, Itose I, Miyazaki M, Kakita N, Higashitani K, Matsubara T, Hiramatsu N, Kasahara A, Takehara T, Hayashi N. Comprehensive immunological analyses of colorectal cancer patients in the phase I/II study of quickly matured dendritic cell vaccine pulsed with carcinoembryonic antigen peptide. Cancer Immunol Immunother. 2011; 60:1565–1575.
crossref
42. Toh HC, Wang WW, Chia WK, Kvistborg P, Sun L, Teo K, Phoon YP, Soe Y, Tan SH, Hee SW, Foo KF, Ong S, Koo WH, Zocca MB, Claesson MH. Clinical benefit of allogeneic melanoma cell lysate-pulsed autologous dendritic cell vaccine in MAGE-positive colorectal cancer patients. Clin Cancer Res. 2009; 15:7726–7736.
crossref
43. Burgdorf SK, Fischer A, Myschetzky PS, Munksgaard SB, Zocca MB, Claesson MH, Rosenberg J. Clinical responses in patients with advanced colorectal cancer to a dendritic cell based vaccine. Oncol Rep. 2008; 20:1305–1311.
crossref
44. Hunyadi J, Andras C, Szabo I, Szanto J, Szluha K, Sipka S, Kovacs P, Kiss A, Szegedi G, Altorjay I, Sapy P, Antal-Szalmas P, Toth L, Fazekas G, Rajnavolgyi E. Autologous dendritic cell based adoptive immunotherapy of patients with colorectal cancer-A phase I-II study. Pathol Oncol Res. 2014; 20:357–365.
crossref
45. Thompson JA, Grunert F, Zimmermann W. Carcinoembryonic antigen gene family: molecular biology and clinical perspectives. J Clin Lab Anal. 1991; 5:344–366.
crossref
46. Kavanagh B, Ko A, Venook A, Margolin K, Zeh H, Lotze M, Schillinger B, Liu W, Lu Y, Mitsky P, Schilling M, Bercovici N, Loudovaris M, Guillermo R, Lee SM, Bender J, Mills B, Fong L. Vaccination of metastatic colorectal cancer patients with matured dendritic cells loaded with multiple major histocompatibility complex class I peptides. J Immunother. 2007; 30:762–772.
crossref
47. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, Simon P, Lotze MT, Yang JC, Seipp CA, Simpson C, Carter C, Bock S, Schwartzentruber D, Wei JP, White DE. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988; 319:1676–1680.
crossref
48. Dudley ME, Wunderlich JR, Yang JC, Hwu P, Schwartzentruber DJ, Topalian SL, Sherry RM, Marincola FM, Leitman SF, Seipp CA, Rogers-Freezer L, Morton KE, Nahvi A, Mavroukakis SA, White DE, Rosenberg SA. A phase I study of nonmyeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes in patients with metastatic melanoma. J Immunother. 2002; 25:243–251.
crossref
49. Gardini A, Ercolani G, Riccobon A, Ravaioli M, Ridolfi L, Flamini E, Ridolfi R, Grazi GL, Cavallari A, Amadori D. Adjuvant, adoptive immunotherapy with tumor infiltrating lymphocytes plus interleukin-2 after radical hepatic resection for colorectal liver metastases: 5-year analysis. J Surg Oncol. 2004; 87:46–52.
crossref
50. Karlsson M, Marits P, Dahl K, Dagoo T, Enerback S, Thorn M, Winqvist O. Pilot study of sentinel-node-based adoptive immunotherapy in advanced colorectal cancer. Ann Surg Oncol. 2010; 17:1747–1757.
crossref
51. Zhen YH, Liu XH, Yang Y, Li B, Tang JL, Zeng QX, Hu J, Zeng XN, Zhang L, Wang ZJ, Li XY, Ge HX, Winqvist O, Hu PS, Xiu J. Phase I/II study of adjuvant immunotherapy with sentinel lymph node T lymphocytes in patients with colorectal cancer. Cancer Immunol Immunother. 2015; 64:1083–1093.
crossref
52. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DA, Feldman SA, Davis JL, Morgan RA, Merino MJ, Sherry RM, Hughes MS, Kammula US, Phan GQ, Lim RM, Wank SA, Restifo NP, Robbins PF, Laurencot CM, Rosenberg SA. T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther. 2011; 19:620–626.
crossref
53. Schmidt-Wolf GD, Negrin RS, Schmidt-Wolf IG. Activated T cells and cytokine-induced CD3+CD56+ killer cells. Ann Hematol. 1997; 74:51–56.
54. Franceschetti M, Pievani A, Borleri G, Vago L, Fleischhauer K, Golay J, Introna M. Cytokine-induced killer cells are terminally differentiated activated CD8 cytotoxic T-EMRA lymphocytes. Exp Hematol. 2009; 37:616–628.e612.
55. Verneris MR, Baker J, Edinger M, Negrin RS. Studies of ex vivo activated and expanded CD8+ NK-T cells in humans and mice. J Clin Immunol. 2002; 22:131–136.
56. Pievani A, Borleri G, Pende D, Moretta L, Rambaldi A, Golay J, Introna M. Dual-functional capability of CD3+CD56+ CIK cells, a T-cell subset that acquires NK function and retains TCR-mediated specific cytotoxicity. Blood. 2011; 118:3301–3310.
crossref
57. Jakel CE, Schmidt-Wolf IG. An update on new adoptive immunotherapy strategies for solid tumors with cytokine-induced killer cells. Expert Opin Biol Ther. 2014; 14:905–916.
crossref
58. Zhu Y, Zhang H, Li Y, Bai J, Liu L, Liu Y, Qu Y, Qu X. Efficacy of postoperative adjuvant transfusion of cytokine-induced killer cells combined with chemotherapy in patients with colorectal cancer. Cancer Immunol Immunother. 2013; 62:1629–1635.
crossref
59. Zhu H, Yang X, Li J, Ren Y, Zhang T, Zhang C, Zhang J, Li J, Pang Y. Immune response, safety, and survival and quality of life outcomes for advanced colorectal cancer patients treated with dendritic cell vaccine and cytokine-induced killer cell therapy. Biomed Res Int. 2014; 2014:603871.
crossref
60. Gao D, Li C, Xie X, Zhao P, Wei X, Sun W, Liu HC, Alexandrou AT, Jones J, Zhao R, Li JJ. Autologous tumor lysate-pulsed dendritic cell immunotherapy with cytokine-induced killer cells improves survival in gastric and colorectal cancer patients. PLoS One. 2014; 9:e93886.
crossref
61. Leibovitz A, Stinson JC, McCombs WB 3rd, McCoy CE, Mazur KC, Mabry ND. Classification of human colorectal adenocarcinoma cell lines. Cancer Res. 1976; 36:4562–4569.
62. Ramirez R, Solana R, Carracedo J, Alonso MC, Pena J. Mechanisms involved in NK resistance induced by interferon-gamma. Cell Immunol. 1992; 140:248–256.
63. Esin E, Yalcin S. Maintenance strategy in metastatic colorectal cancer: A systematic review. Cancer Treat Rev. 2016; 42:82–90.
crossref
64. Ko JJ, Kennecke HF, Lim HJ, Renouf DJ, Gill S, Woods R, Speers C, Cheung WY. Reasons for underuse of adjuvant chemotherapy in elderly patients with stage III colon cancer. Clin Colorectal Cancer. 2015; DOI: 10.1016/j.clcc.2015.09.002.
crossref
65. Morse MA, Niedzwiecki D, Marshall JL, Garrett C, Chang DZ, Aklilu M, Crocenzi TS, Cole DJ, Dessureault S, Hobeika AC, Osada T, Onaitis M, Clary BM, Hsu D, Devi GR, Bulusu A, Annechiarico RP, Chadaram V, Clay TM, Lyerly HK. 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. 2013; 258:879–886.
crossref
66. Morse MA, Clay TM, Hobeika AC, Osada T, Khan S, Chui S, Niedzwiecki D, Panicali D, Schlom J, Lyerly HK. Phase I study of immunization with dendritic cells modified with fowlpox encoding carcinoembryonic antigen and costimulatory molecules. Clin Cancer Res. 2005; 11:3017–3024.
crossref
67. Matsuda K, Tsunoda T, Tanaka H, Umano Y, Tanimura H, Nukaya I, Takesako K, Yamaue H. Enhancement of cytotoxic T-lymphocyte responses in patients with gastrointestinal malignancies following vaccination with CEA peptide-pulsed dendritic cells. Cancer Immunol Immunother. 2004; 53:609–616.
crossref
TOOLS
Similar articles