Purpose: The therapeutic benefit of the cytokine-induced killer (CIK) cells was unknown in the renal cell carcinoma (RCC). This prospectively randomized study was conducted to evaluate the effects of autologous CIK cell immunotherapy in patients with metastatic clear cell RCCs.

Experimental Design: From June 2005 to June 2008, 148 patients with metastatic clear cell RCC were randomized to autologous CIK cell immunotherapy (arm 1, n = 74), or interleukin-2 treatment combination with IFN-α-2a (arm 2, n = 74). The primary endpoint was overall survival (OS) and secondary endpoint was progression-free survival (PFS) evaluated by Kaplan–Meier analyses and treatment HRs with the Cox proportional hazards model.

Results: The 3-year PFS and OS in arm 1 were 18% and 61%, as compared with 12% and 23% in arm 2 (P = 0.031 and <0.001, respectively). The median PFS and OS in arm 1 were significantly longer than those in arm 2 (PFS, 12 vs. 8 months, P = 0.024; OS, 46 vs. 19 months, P < 0.001). Multivariate analyses indicated that the cycle count of CIK cell immunotherapy as a continuous variable was significantly associated with prolonged PFS [HR = 0.88; 95% confidence interval (CI), 0.84-0.93; P < 0.001] and OS (HR = 0.58; 95% CI, 0.48–0.69; P < 0.001) in arm 1.

Conclusion: The data suggested that CIK cell immunotherapy could improve the prognosis of metastatic clear cell RCC, and increased cycle count of CIK cell treatment could further enhance the beneficial effects. Clin Cancer Res; 18(6); 1751–9. ©2012 AACR.

Translational Relevance

Cytokine-induced killer (CIK) cells have the ability to kill tumor cells in vitro and in vivo. However, current data from phase I studies on the anti–renal cell carcinoma (RCC) effects of CIK cells are highly limited, and the therapeutic benefits of CIK cells are unknown in metastatic RCCs. This prospectively randomized study was to evaluate the effects of autologous CIK cell immunotherapy in patients with metastatic clear cell RCCs. One hundred and forty-eight patients with metastatic clear cell RCC were randomized to autologous CIK cell immunotherapy or interleukin-2 treatment combination with IFN-α-2a. Our findings indicate that CIK cell immunotherapy could improve the prognosis of metastatic clear cell RCCs, and increased cycle count of CIK cell treatment could further enhance the beneficial effects.

Metastatic renal cell carcinoma (RCC) has a poor prognosis with a median overall survival (OS) of 12 months and a 5-year survival rate of less than 10% (1). Metastatic RCC seems to be resistant to conventional therapy regimens such as chemotherapy, hormone therapy, and radiotherapy with an objective response rate of less than 10% (1). The most successful therapy for metastatic RCC has been single-agent, high-dose interleukin (IL)-2 with durable complete remissions in a small percentage of patients. This is at the expense of considerable toxicity and thus careful patient selection by clinical and other criteria is appropriate (2). Prior attempts to improve IL-2 clinical outcome with the addition of other agents have shown a very limited success (3–5). New targeted agents, such as sunitinib, sorafenib, temsirolimus, and bevacizumab, have been developed and are used as a first-line therapy in many centers. Although these agents represent a major advance in the treatment of this disease, they are palliative treatments and rarely produce durable complete remissions (6–9). These limited successes indicate that further efforts are needed to improve the current therapeutic modalities and to explore novel therapies for RCCs to improve patient care and increase survival (10, 11).

Immunotherapy has recently become the fourth important treatment modality for malignant tumors, ranked after surgery, radiotherapy, and chemotherapy (12–14). A number of adoptive immunotherapies using various killer cells have been reported, including lymphokine-activated killer cells (LAK), tumor-infiltrating lymphocytes (TIL), and anti-CD3 monoclonal antibody–induced killer cells (15–17). However, the therapeutic efficacy is limited because of their low antitumor activities (10). At present, cytokine-induced killer (CIK) cells have been recognized as a new type of antitumor effector cells, which can proliferate rapidly in vitro, with stronger antitumor activity and broader spectrum of targeted tumor than other reported antitumor effector cells (13). Moreover, CIK cells can regulate and generally enhance the immune functions in patients with cancer (18). Our previous phase I study showed the feasibility and low toxicity of CIK cell immunotherapy on patients with cancer (19). However, current data from phase I studies on the anti-RCC effects of CIK cells are highly limited (19–21), and the therapeutic benefits of CIK cells are unknown in metastatic RCCs. The purpose of this prospectively randomized study was to evaluate the clinical efficacy of CIK cell immunotherapy in patients with metastatic clear cell RCCs.

Patients' selection

Between June 2005 and June 2008, 148 patients with metastatic clear cell RCCs were treated in this randomized clinical trial (Table 1). Criteria for entry onto this study were histologically confirmed progressive and irresectable metastatic clear cell RCCs, expected survival duration of more than 3 months, Karnofsky performance status (KPS) more than 40%, age between 18 and 80 years, serum bilirubin and creatinine less than 1.25 of the upper normal limit, and free of congestive heart failure, severe coronary artery disease, cardiac arrhythmias, HIV infection, chronic active hepatitis, and concomitant corticosteroid therapy. In all patients treated, no chemotherapy or immunomodulatory treatment had been conducted during the previous 4 weeks. Pregnant and lactating women were excluded.

Table 1.

Distributions of demographic and clinical characteristics of patients in arms 1 and 2

Demographic and clinical featuresArm 1Arm 2P
No. of patients 74 74 NA 
Sex   0.831 
Male (n60 61  
Female (n14 13  
Age 
Median (range), y 59 (35–78) 60 (40–79) 0.742 
60 y   0.741 
 ≥60 (n33 35  
 <60 (n41 39  
KPS   0.503 
<80 (n46 42  
≥80 (n28 32  
Hemoglobin   0.452 
<LLN (n17 21  
≥LLN (n57 53  
LDH   0.745 
>1.5 × ULN (n 
≤1.5 × ULN (n68 70  
Serum corrected calcium   0.772 
>ULN (n 
≤ULN (n68 67  
Time from initial diagnosis to current therapy   0.508 
<1 y (n35 31  
>1 y (n39 43  
Risk category   0.826 
Favorable risk (n19 17  
Intermediate risk (n39 38  
Poor risk (n16 19  
Neutrophils   0.547 
>ULN (n 
≤ULN (n69 67  
Platelets   0.711 
>ULN (n19 21  
≤ULN (n55 53  
Sites of metastatic disease   0.638 
Peripheral lymph nodes (n 
Lung (n51 53  
Bone (n21 20  
Liver (n11  
Retroperitoneum (n 
Pancreas (n 
Brain (n 
Others (n 
No. of metastases   0.487 
>1 (n27 23  
<1 (n47 51  
Nephrectomy   0.545 
No (n17 14  
Yes (n57 60  
Prior therapy 
Yes/no   0.317 
 Yes (n28 34  
 No (n46 40  
Treatment category   0.744 
 Radiation therapy (n12 16  
 Chemotherapy (n 
 Immunotherapy (n11 17  
 Traditional Chinese drug (n.)  
 Renal artery/veins interventional (n 
Subsequent therapy 
Yes/no   0.099 
 Yes (n35 45  
 No (n39 29  
Treatment category   0.830 
 Radiotherapy (n11 20  
 Chemotherapy (n18 22  
 Target therapy (n 
 Traditional Chinese drug (n14 17  
Demographic and clinical featuresArm 1Arm 2P
No. of patients 74 74 NA 
Sex   0.831 
Male (n60 61  
Female (n14 13  
Age 
Median (range), y 59 (35–78) 60 (40–79) 0.742 
60 y   0.741 
 ≥60 (n33 35  
 <60 (n41 39  
KPS   0.503 
<80 (n46 42  
≥80 (n28 32  
Hemoglobin   0.452 
<LLN (n17 21  
≥LLN (n57 53  
LDH   0.745 
>1.5 × ULN (n 
≤1.5 × ULN (n68 70  
Serum corrected calcium   0.772 
>ULN (n 
≤ULN (n68 67  
Time from initial diagnosis to current therapy   0.508 
<1 y (n35 31  
>1 y (n39 43  
Risk category   0.826 
Favorable risk (n19 17  
Intermediate risk (n39 38  
Poor risk (n16 19  
Neutrophils   0.547 
>ULN (n 
≤ULN (n69 67  
Platelets   0.711 
>ULN (n19 21  
≤ULN (n55 53  
Sites of metastatic disease   0.638 
Peripheral lymph nodes (n 
Lung (n51 53  
Bone (n21 20  
Liver (n11  
Retroperitoneum (n 
Pancreas (n 
Brain (n 
Others (n 
No. of metastases   0.487 
>1 (n27 23  
<1 (n47 51  
Nephrectomy   0.545 
No (n17 14  
Yes (n57 60  
Prior therapy 
Yes/no   0.317 
 Yes (n28 34  
 No (n46 40  
Treatment category   0.744 
 Radiation therapy (n12 16  
 Chemotherapy (n 
 Immunotherapy (n11 17  
 Traditional Chinese drug (n.)  
 Renal artery/veins interventional (n 
Subsequent therapy 
Yes/no   0.099 
 Yes (n35 45  
 No (n39 29  
Treatment category   0.830 
 Radiotherapy (n11 20  
 Chemotherapy (n18 22  
 Target therapy (n 
 Traditional Chinese drug (n14 17  

Abbreviations: LLN, lower limit of normal; NA, not applicable; ULN, upper limit of normal.

This study was approved by the State Food and Drug Administration, China (2006L01023), and by the Ethical Committee of Cancer Hospital of Tianjin Medical University, Tianjin, China, according to the guidelines of the Declaration of Helsinki. Informed consent was obtained from all subjects before their entry onto the study. Follow-ups of all patients were started from June 1, 2005, and ended on June 1, 2011. Median follow-up of all patients was 27 months (range, 3–72 months).

Study design

Patients were randomly assigned to receive intravenous infusion autologous CIK cells (iv-CIKs; arm 1) or subcutaneous injection human IL-2 (sc-IL-2) in combination with subcutaneous injection human IFN-α-2a (sc-IFN-α-2a; arm 2). The major endpoint was OS and secondary endpoint was progression-free survival (PFS) in this trial. The sample size calculation assumed a 20% improvement in OS at 3 years with a one-sided α-risk of 5% and a β-risk of 20%. OS rates at 3 years were estimated to be 45% and 25% in arms 1 and 2, respectively. Under these assumptions, 140 patients would be required to detect a significant difference between groups.

Randomization was stratified by using a block method. Treatment allocation was done through a specific web site. An independent review committee evaluated the quality of the study, especially with respect to independent radiographic assessment, data monitoring, and statistical analysis.

Treatments

As shown in Fig. 1, patients received iv-CIKs at days 15 and 16 per cycle in arm 1 and received cycle treatment once every month. Patients without disease progression were eligible for maintenance treatment. Patients received a median of 97 × 108 (range, 76 × 108–114 × 108) autologous CIK cells every cycle.

Figure 1.

Trial and treatment summary. CR, complete response; PR, partial response; SD, stable disease.

Figure 1.

Trial and treatment summary. CR, complete response; PR, partial response; SD, stable disease.

Close modal

In arm 2, sc-IL-2 (Proleukin, Chiron) and sc-IFN-α-2a (Roferon, Hoffmann-La Roche) were administered as previously reported in the CRECY study (22). IL-2 was administered to patients at 10 × 106 IU/m2/d on days 1, 3, and 5, weeks 1 to 4. IFN-α-2a was administered at 3 × 106 IU/m2/d on days 2, 4, and 6, weeks 1 to 4. After 2 weeks of rest, an identical treatment with IL-2 combination with IFN-α-2a was administered. Six-week treatment cycles were repeated for up to 1 year unless progression of disease occurred.

Detailed recommendations for dose modifications were given to the investigators. In summary, all toxic events judged as grade III for intensity and prolonged over 2 weeks, or events of grade IV intensity, could justify permanent treatment discontinuation. Concomitant medication was given as needed to control adverse effects. Patients with progressive disease in arms 1 or 2 left study to receive subsequent individual care. Crossover between treatment arms was not allowed. Reevaluation of the patient's tumor status was conducted every 2 months. In arm 1, one (1.4%) patient was delayed treatment because of grade III fatigue. There was no patient who received dose modification and no patient failing to fulfill the CIK cell immunotherapy in arm 1. The median cycle of CIK cell immunotherapy was 10 cycles (range, 2–35 cycles). In arm 2, 14 (19%) patients did not complete cycle 1 because of early progression before the first response evaluation (6%), intolerance (10%), or both early disease progression and intolerance (3%). Beside these, 4 (5.4%) patients delayed treatment and 6 (8.1%) patients received dose modification. Patients in arm 2 received a median of 2.5 six-week cycles (range, 1–8 cycles). All patients were seen at regular biweekly intervals by oncologic specialists; additional care was provided whenever needed.

Assessment of response and toxicity

Risk category of patients was preformed according to the Memorial Sloan-Kettering Cancer Center (MSKCC, New York) criteria (23), Tumor response was classified in accordance with the National Cancer Institute (Bethesda, MD) Response Evaluation Criteria in Solid Tumors (RECIST; ref. 24). Patients were assessed serially using computed tomography of chest, abdomen, pelvis, and brain, and technetium bone scan. Tumor evaluation was done 2 months after treatment start. Adverse events were evaluated according to World Health Organization (WHO) criteria.

CIK cells preparation

CIK cells were prepared as described in our previous studies (19, 25). Briefly, peripheral blood mononuclear cells (PBMC) were collected from the patients with RCCs using a Cobe Spectra Apheresis System (CaridianBCT), and cultured in AIM-V medium (Invitrogen) containing 50 ng/mL anti-CD3 antibody (e-Bioscience), 100 U/mL recombinant human IL-1α (e-Bioscience), and 1,000 U/mL IFN-γ (PeproTech), at 37°C with 5% CO2 for 24 hours. Then, 300 U/mL recombinant human IL-2 (rhIL-2; Proleukin) was added to the media. The medium was replaced by the fresh IL-2 and IFN-γ–containing medium was replaced every 5 days. At day 14, CIK cells were harvested and analyzed for phenotype and cytotoxicity. All products were free of bacterial, mycoplasma, or fungal contamination. The endotoxin was less than 5 EU.

Detecting the phenotype of CIK cells

Phenotypes of untreated PBMCs and autologous CIK cells from 29 patients selected randomly from the 74 patients in arm 1 were detected by two-color fluorescence as described in our previous studies (19, 25). Briefly, according to the routine method, 5 × 105 CIK cells were resuspended in 20 μL of PBS containing 2% newborn calf serum and 1% sodium azide and then labeled with anti-CD3-FITC/anti-CD56-RPE (Dako), CD3-FITC (fluorescein isothiocyanate), CD4-RPE, CD8-RPE, CD45RO, and CD4-FITC/CD25-PE (BD Bioscience). The cell population was analyzed using flow cytometry (BD Aria).

Detecting cytotoxicity of CIK cells

The cytotoxicity of CIK cells from 29 patients selected randomly from the 74 patients in arm 1 was detected as described in our previous studies (19, 25). Briefly, the target cells used for this assay included K562, CALU-6, 823, MCF-7, 786-O, and SK-RC-42. These cell lines were obtained from the American Type Culture Collection. Target cells (1 × 105 cells/mL) were incubated for 4 hours in triplicate sets with effector cells (CIK cells) at an effector-to-target cell ratio of 50:1. At the end of incubation, 50 μL culture supernatant was transferred to a new, flat 96-well plate, and incubated with 50 μL lactate dehydrogenase (LDH) substrate mixture (for detection of LDH released upon cell lysis) at room temperature for 30 minutes in dark. Then, 50 μL stop solution was added to each well. Absorbance was measured at 490 nm using a 96-well plate reader. Killing efficiency was calculated as: % killing efficiency = [(experimental counts − effector spontaneous counts − target spontaneous counts)/(target maximal counts − target spontaneous counts)] × 100.

Statistical methods

OS was calculated from initiation of treatment to death, and patients alive were censored at the time of last contact. PFS was calculated from initiation of treatment until first progression, and patients alive in stable state were censored at the time of last contact. The χ2 test and Fisher exact test were used for binary variable comparisons. The Mann–Whitney U test was used for median comparisons. Distributions of survival time and rate were estimated by using the Kaplan–Meier method; median survival time and 3-year survival rates along with 95% confidence intervals (CI) were reported. Associations between survival and potential prognostic factors were assessed using the log-rank test in univariable analyses. The Cox proportional hazards model was undertaken in multivariable analyses by using the forward-LR (likelihood ratio) method with a significance level of 0.15 for entering and removing variables. In univariate evaluations of the prognostic impact of continuous variable (the cycle count of CIK cell treatment), the optimal cutoff point was determined using the receiver operating characteristic curve method. A P value less than 0.05 using two-sided tests indicates statistical significance. All calculations were conducted using the SPSS 16.0 Software.

Patient characteristics

Seventy-four patients were randomized to the CIK cell therapy (arm 1) and 74 patients to the IL-2 plus IFN-α-2a treatment (arm 2). The characteristics of patients in the 2 groups are detailed in Table 1; there were no significant differences between the 2 groups.

Amplification of CIK cells in vitro

The median count of untreated PBMCs of all patients in arm 1 was 21 × 108 (range, 7 × 108–32 × 108) per cycle. The median count of autologous CIK cells after 14 days of amplification could reach 97 × 108 (range, 76 × 108–114 × 108) per cycle. On the basis of trypan blue staining, the cellular vitality reached 95% or even higher.

Phenotypic analysis of CIK cells

Phenotypic analysis of autologous CIK cells in 29 patients of arm 1 before culture and after 14 days of culture showed that percentages of CD3+, CD3+ CD4+, CD3+ CD8+, CD3+ CD56+, and CD25+ cell subsets increased from 48.41% ± 6.19%, 28.07% ± 4.76%, 19.00% ± 5.54%, 3.65% ± 1.41%, and 13.11% ± 3.11% to 81.06% ± 9.22%, 42.70% ± 6.18%, 36.41% ± 5.19%, 18.21% ± 4.73%, and 33.13% ± 6.87%, respectively, with P values <0.01. But percentages of CD3/16+/56+, CD14+, and CD20+ cell subsets decreased from 14.23% ± 3.34%, 17.44% ± 5.06%, and 12.11% ± 4.14% to 7.18% ± 2.01%, 6.17% ± 1.89%, and 8.14% ± 2.42%, respectively, with P values <0.05.

Cytotoxicity of CIK cells against tumor cells in vitro

Autologous CIK cells from the 29 patients in arm 1 were mixed with various types of tumor cell lines at an effector-to-target ratio of 50:1. Mean values of cytotoxicity of CIK cells against K562, CALU-6, 823, and MCF-7 were 45.30% ± 7.95%, 28.51% ± 6.22%, 34.11% ± 8.27%, and 25.06% ± 6.61%, respectively. In addition, the cytotoxic activity of CIK cells from patients with metastatic RCCs was investigated against 2 human renal carcinoma cell lines 786-O and SK-RC-42. At an effector-to-target ratio of 50:1, CIK cells killed 786-O and SK-RC-42 cells with mean percentage lysis of 35.41% ± 6.11% and 32.17% ± 5.30%, respectively.

Treatment response

Thirteen patients (18%) in arm 1 achieved a complete response and 26 patients (35%) had a partial remission. The overall objective response rate was 53% (95% CI, 45%–61%). Twenty-five patients (34%) showed disease stabilization and 10 patients (14%) exhibited continuous disease progression despite therapy. In arm 2, there were 5 complete responders (7%) and 15 partial responders (20%), with an overall objective response rate of 27% (95% CI, 17%–37%). Twenty-five patients (34%) had disease stabilization, and in 29 patients (39%), a continuous disease progression was observed.

Prognosis of patients in the two groups

The 3-year PFS rate in arm 1 (18%; 95% CI, 10%–26%) was significantly higher than that in arm 2 (12%; 95% CI, 6%–18%; P = 0.031). The 3-year OS rate in arm 1 (61%; 95% CI, 52%–70%) was also significantly higher than that in arm 2 (23%; 95% CI, 16%–30%; P < 0.001). The median PFS in arm 1 (12 months; 95% CI, 10–14 months) was significantly longer than that in arm 2 (8 months; 95% CI, 4–12 months; P = 0.024; Fig. 2A). The median OS in arm 1 (46 months; 95% CI, 36–56 months) was also significantly longer than that in arm 2 (19 months; 95% CI, 13–25 months; P < 0.001; Fig. 2B).

Figure 2.

Prognosis of patients in arms 1 and 2. A, PFS; B, OS.

Figure 2.

Prognosis of patients in arms 1 and 2. A, PFS; B, OS.

Close modal

Prognostic factors of patients in arm 1

The cycle count of CIK cell treatment significantly improved the PFS and OS of patients when analyzed as a continuous variable in the univariate analysis (P < 0.001 and <0.001, respectively) and in the multivariate analysis after adjustment for sex, age, KPS, hemoglobin, neutrophils, platelets, LDH, serum corrected calcium, time from initial diagnosis to current therapy, number of metastatic disease, nephrectomy, prior and poststudy therapies (HR = 0.88, 95% CI, 0.84–0.93, P < 0.001; and HR = 0.58, 95% CI, 0.48–0.69, P < 0.001; respectively). The optimal cutoff point of the cycle count was 7 cycles. All 14 potential predictive covariates with their univariate analyses are presented in Table 2. KPS less than 80, no prior nephrectomy, time from initial diagnosis to current treatment of less than 1 year, number of metastases greater than 1, anemia, hypercalcemia, neutrophils, and elevated LDH were significantly associated with poor prognosis. The cycle count of CIK treatments ≥7 cycles was significantly associated with good prognosis. In the multivariate analysis, KPS less than 80, time from initial diagnosis to current treatment of less than 1 year, number of metastases greater than 1, anemia, hypercalcemia, and elevated LDH were independent poor prognostic factors. The cycle count of CIK treatments ≥7 cycles was an independent good prognostic factor (Table 3).

Table 2.

Univariate analysis of 74 patients' demographic and clinical characteristics and survival in the CIK group

ParameterMedian PFS, moLog-rank PMedian OS, moLog-rank P
Sex  0.485  0.093 
 Male 12  45  
 Female 15  52  
Age, y  0.284  0.079 
 ≥60 12  41  
 <60 15  50  
Cycle count of CIK treatment  <0.001  <0.001 
 ≥7 cycles 18  53  
 <7 cycles  17  
Nephrectomy  0.042  0.058 
 Yes 16  46  
 No  27  
Prior therapy  0.102  0.183 
 Yes 12  40  
 No 15  46  
Subsequent therapy  0.147  0.151 
 Yes 12  39  
 No 13  48  
KPS  0.017  0.003 
 ≥80 16  50  
 <80  25  
Hemoglobin  0.015  0.007 
 ≥LLN 17  48  
 <LLN  19  
LDH  0.021  <0.001 
 >1.5 × ULN  15  
 ≤1.5 × ULN 15  47  
Serum corrected calcium  0.013  0.005 
 >ULN  13  
 ≤ ULN 15  47  
Time from initial diagnosis to current therapy, y  0.016  0.008 
 <1 10  42  
 >1 14  55  
Neutrophils  0.009  0.014 
 >ULN  20  
 ≤ULN 15  46  
Platelets  0.029  0.068 
 >ULN 10  39  
 ≤ULN 14  50  
No. of metastases  0.040  <0.001 
 >1 11  20  
 =1 15  50  
ParameterMedian PFS, moLog-rank PMedian OS, moLog-rank P
Sex  0.485  0.093 
 Male 12  45  
 Female 15  52  
Age, y  0.284  0.079 
 ≥60 12  41  
 <60 15  50  
Cycle count of CIK treatment  <0.001  <0.001 
 ≥7 cycles 18  53  
 <7 cycles  17  
Nephrectomy  0.042  0.058 
 Yes 16  46  
 No  27  
Prior therapy  0.102  0.183 
 Yes 12  40  
 No 15  46  
Subsequent therapy  0.147  0.151 
 Yes 12  39  
 No 13  48  
KPS  0.017  0.003 
 ≥80 16  50  
 <80  25  
Hemoglobin  0.015  0.007 
 ≥LLN 17  48  
 <LLN  19  
LDH  0.021  <0.001 
 >1.5 × ULN  15  
 ≤1.5 × ULN 15  47  
Serum corrected calcium  0.013  0.005 
 >ULN  13  
 ≤ ULN 15  47  
Time from initial diagnosis to current therapy, y  0.016  0.008 
 <1 10  42  
 >1 14  55  
Neutrophils  0.009  0.014 
 >ULN  20  
 ≤ULN 15  46  
Platelets  0.029  0.068 
 >ULN 10  39  
 ≤ULN 14  50  
No. of metastases  0.040  <0.001 
 >1 11  20  
 =1 15  50  

Abbreviations: LLN, lower limit of normal; ULN, upper limit of normal.

Table 3.

Multivariable analysis of 74 patients' demographic and clinical characteristics and survival in the CIK group

PFSOS
ParameterHR (95% CI)PHR (95% CI)P
CIK treatments ≥ 7 cycles 0.22 (0.13–0.39) <0.001 0.01 (0.01–0.02) <0.001 
KPS < 80 1.35 (1.17–2.51) 0.037 2.39 (1.87–4.91) 0.012 
Hemoglobin < LLN 1.47 (1.21–2.79) 0.025 4.59 (1.65–10.87) 0.004 
LDH > 1.5×ULN 1.29 (1.10–2.08) 0.023 2.87 (1.40–5.86) 0.004 
Calcium > ULN 2.26 (1.33–4.79) 0.011 6.16 (3.44–11.97) <0.001 
Time from diagnosis to therapy < 1 y 1.72 (1.12–2.54) 0.034 4.19 (1.82–9.64) <0.001 
No. of metastases > 1 1.18 (1.03–2.18) 0.074 1.82 (1.26–2.89) 0.046 
Neutrophils > ULN 1.21 (0.86–2.07) 0.099 1.71 (0.81–3.12) 0.081 
Platelets > ULN 1.15 (0.79–2.26) 0.092 1.84 (0.88–3.34) 0.087 
PFSOS
ParameterHR (95% CI)PHR (95% CI)P
CIK treatments ≥ 7 cycles 0.22 (0.13–0.39) <0.001 0.01 (0.01–0.02) <0.001 
KPS < 80 1.35 (1.17–2.51) 0.037 2.39 (1.87–4.91) 0.012 
Hemoglobin < LLN 1.47 (1.21–2.79) 0.025 4.59 (1.65–10.87) 0.004 
LDH > 1.5×ULN 1.29 (1.10–2.08) 0.023 2.87 (1.40–5.86) 0.004 
Calcium > ULN 2.26 (1.33–4.79) 0.011 6.16 (3.44–11.97) <0.001 
Time from diagnosis to therapy < 1 y 1.72 (1.12–2.54) 0.034 4.19 (1.82–9.64) <0.001 
No. of metastases > 1 1.18 (1.03–2.18) 0.074 1.82 (1.26–2.89) 0.046 
Neutrophils > ULN 1.21 (0.86–2.07) 0.099 1.71 (0.81–3.12) 0.081 
Platelets > ULN 1.15 (0.79–2.26) 0.092 1.84 (0.88–3.34) 0.087 

Abbreviations: LLN, lower limit of normal; ULN, upper limit of normal.

Treatment toxicity

The distributions of side effects in the 2 groups are shown in Table 4. No toxic death was observed. The toxicities in arm 2 were more frequent and more serious than that in arm 1. No patient in arm 1 failed to fulfill the CIK immunotherapy. There were no grade III–IV cell-related toxicities, and common grade I–II toxicities consisted of transient fever, chills, fatigue, headache, and anemia in arm 1.

Table 4.

Distributions of systemic maximum toxicity in the two groups

Arm 1 (%)Arm 2 (%)
Side effects (WHO criteria)Grade I–IIGrade III–IVGrade I–IIGrade III–IV
Fever 21 49 
Chills 10 29 
Fatigue 19 41 
Nausea or vomiting 13 
Diarrhea 10 
Respiratory distress 21 
Skin or allergies 15 
Mucositis 12 
Hypotension 
Arrhythmias 
Headache 10 
Paresthesias 
Fluid retention or edema 
Leukopenia 
Thrombocytopenia 
Anemia 15 
Arm 1 (%)Arm 2 (%)
Side effects (WHO criteria)Grade I–IIGrade III–IVGrade I–IIGrade III–IV
Fever 21 49 
Chills 10 29 
Fatigue 19 41 
Nausea or vomiting 13 
Diarrhea 10 
Respiratory distress 21 
Skin or allergies 15 
Mucositis 12 
Hypotension 
Arrhythmias 
Headache 10 
Paresthesias 
Fluid retention or edema 
Leukopenia 
Thrombocytopenia 
Anemia 15 

Treatment of RCCs, especially metastatic RCCs, confronts a great dilemma in clinical practice. Although there are a number of therapeutic options available such as the immunoregulating cytokines and the new antiangiogenic targeted agents at present, these are commonly toxic and rarely produce durable complete remissions. The recent considerable success of cell immunotherapy in melanoma warrants further efforts to apply this treatment to other cancers including RCCs. Two conventional adoptive cell immunotherapy approaches are LAK cells and TILs (10). LAK cell in combination with IL-2 has been extensively studied and was shown to be heterogeneous and capable of killing both allogeneic and autologous tumors (26). The activity of LAK cells was mainly mediated by natural killer (NK) cells and also indirectly by MHC unrestricted T cells (26). TILs represent a part of the host immune response to human malignancy and contain an enriched population of cells with both cytotoxic and helper functions that are reactive to the autologous tumor (27). The majority of TILs expanded by IL-2 are composed of both CD3+/CD4+ and CD3+/CD8+ T cells (27). In addition, TILs have been shown to contain antigen-specific as well as nonspecific cytotoxic lymphocytes (28). These cell immunotherapies get considerable success on the treatment of melanoma which is regarded as an immunosensitive tumor. Although RCC is an immunosensitive cancer, similar attempts in metastatic RCCs have shown a very limited success (10). CIK cells are a novel population of immune effector cells and are activated T cells with NK cell properties that can be expanded in vitro in presence of rhIL-2, starting from PBMCs stimulated by IFN-γ and anti-CD3 antibody (18). CIK cells express CD3 and CD56 as well as the NKG2D antigen and show MHC-unrestricted cytotoxicity toward neoplastic but not normal targets (18, 29, 30). CIK cells express several chemokine receptors and are shown to migrate to the tumor site after intravenous administration in in vivo models (31, 32). At the tumor site, CIK cells can exert their cytotoxic activity and control tumor growth. CIK cells can proliferate rapidly in vitro, with stronger antitumor activity, broader target tumor spectrum, and lower adverse effect than other reported antitumor effector cells (13, 18). Moreover, CIK cells can regulate and enhance the immune function in patients with cancer (18). Their ease of production in vitro and antitumor potential have made them suitable candidates for cell therapy regimens in solid and hematopoietic tumor treatments. Indeed both autologous and allogeneic CIK cells have been used in phase I/II clinical trials for the treatment of various tumor types. In these trials, they have shown limited toxicity in vivo and evidence of antitumor activity (19–21, 25, 33). However, current knowledge on the anti-RCC effects of CIK cells in phase I studies is highly limited (19–21), and the therapeutic benefit of CIK cells is unknown in metastatic RCCs. To our knowledge, the present report is the largest prognostic study in metastatic RCCs treated with CIK cell immunotherapy. We have shown for the first time that CIK cell treatment significantly improves the prognosis of metastatic RCCs. Heng and colleagues reported that the median OS of metastatic RCCs treated with VEGF-targeted therapy was 28 months (34). In this study, the median OS in patients with metastatic clear cell RCCs treated with CIK cell immunotherapy was 46 months. These data suggest that CIK cell immunotherapy could improve the prognosis of patients with metastatic RCCs.

In multivariate analysis, the cycle count of CIK cell treatment is significantly associated with prognosis in the arm 1, when viewed as a continuous variable. The optimal cutoff point of the cycle count was determined to be 7 cycles. The prognosis of patients who received CIK cell treatments for ≥7 cycles was significant better than that of patients who received <7 cycles of the treatments. The median time from the first to seventh treatment was 9 months (range, 8–12 months). Previous studies have shown that the minimal time for immunotherapy to display an effect in patients with cancer is about 8 months (35, 36). These data indicate that the time CIK cell immunotherapy starts to display an effect in metastatic RCCs is approximately 9 months, and the maintenance treatments are required after the initial effect is observed for maximal benefits. However, the preferred length for maintenance treatments is still unclear and needs further investigations.

In conclusion, we have revealed, for the first time, the relationship between the CIK cell immunotherapy and the prognosis of metastatic RCCs. Our study has indicated that CIK cell treatment could improve the prognosis of metastatic clear cell RCCs, and increased frequency of CIK cell immunotherapy could result in additional benefits.

No potential conflicts of interest were disclosed.

L. Liu, H. Li, J. Yu, and S. Wei contributed to the conception and design of the study. L. Liu and H. Li contributed to the development of methodology and writing, review, and/or revision of the manuscript. L. Liu, W. Zhang, and H. Li contributed to the acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.). L. Liu and X. Qi contributed to the analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis) and fulfilled the statistical analysis. L. Liu provided administrative, technical, or material support (i.e., reporting or organizing data, constructing databases) and conducted study supervision. X. Hao and X. Ren were the principal investigators and took the primary responsibility for the manuscript. W. Zhang, X. Ren, and X. Hao recruited the patients. L. Liu, H. Li, and J. Yu conducted the laboratory work for this study. L. Liu, X. Ren, and S. Wei wrote the manuscript.

This work was supported partially by the National Natural Science Funds (No. 30872986 and No. 30901754) and by the Tianjin Key Natural Science Funds (No. 09JCZDJC20400).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Garcia
JA
,
Rini
BI
. 
Recent progress in the management of advanced renal cell carcinoma
.
CA Cancer J Clin
2007
;
57
:
112
5
.
2.
McDermott
DF
. 
Update on the application of interleukin-2 in the treatment of renal cell carcinoma
.
Clin Cancer Res
2007
;
13
:
716s
20s
.
3.
Atzpodien
J
,
Kirchner
H
,
Jonas
U
,
Bergmann
L
,
Schott
H
,
Heynemann
H
, et al
Interleukin-2- and interferon alfa-2a-based immunochemotherapy in advanced renal cell carcinoma: a Prospectively Randomized Trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN)
.
J Clin Oncol
2004
;
22
:
1188
94
.
4.
Yang
JC
,
Haworth
L
,
Sherry
RM
,
Hwu
P
,
Schwartzentruber
DJ
,
Topalian
SL
, et al
A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer
.
N Engl J Med
2003
;
349
:
427
34
.
5.
Négrier
S
,
Perol
D
,
Ravaud
A
,
Bay
JO
,
Oudard
S
,
Chabaud
S
, et al
Randomized study of intravenous versus subcutaneous interleukin-2, and IFNalpha in patients with good prognosis metastatic renal cancer
.
Clin Cancer Res
2008
;
14
:
5907
12
.
6.
Motzer
RJ
,
Michaelson
MD
,
Redman
BG
,
Hudes
GR
,
Wilding
G
,
Figlin
RA
, et al
Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2006
;
24
:
16
24
.
7.
Ahmad
T
,
Eisen
T
. 
Kinase inhibition with BAY 43-9006 in renal cell carcinoma
.
Clin Cancer Res
2004
;
10
:
6388s
92s
.
8.
Atkins
MB
,
Hidalgo
M
,
Stadler
WM
,
Logan
TF
,
Dutcher
JP
,
Hudes
GR
, et al
Randomized phase II study of multiple dose levels of CCI-779, a novel mammalian target of rapamycin kinase inhibitor, in patients with advanced refractory renal cell carcinoma
.
J Clin Oncol
2004
;
22
:
909
18
.
9.
Ko
JS
,
Zea
AH
,
Rini
BI
,
Ireland
JL
,
Elson
P
,
Cohen
P
, et al
Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients
.
Clin Cancer Res
2009
;
15
:
2148
57
.
10.
Shablak
A
,
Hawkins
RE
,
Rothwell
DG
,
Elkord
E
. 
T cell-based immunotherapy of metastatic renal cell carcinoma: modest success and future perspective
.
Clin Cancer Res
2009
;
15
:
6503
10
.
11.
Rini
BI
. 
New strategies in kidney cancer: therapeutic advances through understanding the molecular basis of response and resistance
.
Clin Cancer Res
2010
;
16
:
1348
54
.
12.
Dougan
M
,
Dranoff
G
. 
Immune therapy for cancer
.
Annu Rev Immunol
2009
;
27
:
83
117
.
13.
Hontscha
C
,
Borck
Y
,
Zhou
H
,
Messmer
D
,
Schmidt-Wolf
IG
. 
Clinical trials on CIK cells: first report of the international registry on CIK cells (IRCC)
.
J Cancer Res Clin Oncol
2011
;
137
:
305
10
.
14.
Schwaab
T
,
Schwarzer
A
,
Wolf
B
,
Crocenzi
TS
,
Seigne
JD
,
Crosby
NA
, et al
Clinical and immunologic effects of intranodal autologous tumor lysate-dendritic cell vaccine with Aldesleukin (Interleukin 2) and IFN-{alpha}2a therapy in metastatic renal cell carcinoma patients
.
Clin Cancer Res
2009
;
15
:
4986
92
.
15.
Rosenberg
S
. 
Lymphokine-activated killer cells: a new approach to immunotherapy of cancer
.
J Natl Cancer Inst
1985
;
75
:
595
603
.
16.
Rosenberg
SA
,
Spiess
P
,
Lafreniere
R
. 
A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes
.
Science
1986
;
233
:
1318
21
.
17.
Yun
YS
,
Hargrove
ME
,
Ting
CC
. 
In vivo antitumor activity of anti-CD3-induced activated killer cells
.
Cancer Res
1989
;
49
:
4770
4
.
18.
Schmidt-Wolf
IG
,
Lefterova
P
,
Mehta
BA
,
Fernandez
LP
,
Huhn
D
,
Blume
KG
, et al
Phenotypic characterization and identification of effector cells involved in tumor cell recognition of cytokine-induced killer cells
.
Exp Hematol
1993
;
21
:
1673
9
.
19.
Ren
X
,
Yu
J
,
Liu
H
,
Zhang
P
,
An
X
,
Zhang
N
, et al
Th1 bias in PBMC induced by multicycles of auto-CIKs infusion in malignant solid tumor patients
.
Cancer Biother Radiopharm
2006
;
21
:
22
33
.
20.
Su
X
,
Zhang
L
,
Jin
L
,
Ye
J
,
Guan
Z
,
Chen
R
, et al
Immunotherapy with cytokine-induced killer cells in metastatic renal cell carcinoma
.
Cancer Biother Radiopharm
2010
;
25
:
465
70
.
21.
Schmidt-Wolf
IG
,
Finke
S
,
Trojaneck
B
,
Denkena
A
,
Lefterova
P
,
Schwella
N
, et al
Phase I clinical study applying autologous immunological effector cells transfected with the interleukin-2 gene in patients with metastatic renal cancer, colorectal cancer and lymphoma
.
Br J Cancer
1999
;
81
:
1009
16
.
22.
Negrier
S
,
Escudier
B
,
Lasset
C
,
Douillard
JY
,
Savary
J
,
Chevreau
C
, et al
Recombinant human interleukin-2, recombinant human interferon α-2a, or both in metastatic renal-cell carcinoma
.
N Engl J Med
1998
;
338
:
1272
8
.
23.
Motzer
RJ
,
Bacik
J
,
Murphy
BA
,
Russo
P
,
Mazumdar
M
. 
Interferon alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma
.
J Clin Oncol
2002
;
20
:
289
96
.
24.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
,
Wanders
J
,
Kaplan
RS
,
Rubinstein
L
, et al
New guidelines to evaluate the response to treatment in solid tumors (RECIST Guidelines)
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
25.
Li
H
,
Wang
C
,
Yu
J
,
Cao
S
,
Wei
F
,
Zhang
W
, et al
Dendritic cell-activated cytokine-induced killer cells enhance the anti-tumor effect of chemotherapy on non-small cell lung cancer in patients after surgery
.
Cytotherapy
2009
;
11
:
1076
83
.
26.
Grimm
EA
,
Mazumder
A
,
Zhang
HZ
,
Rosenberg
SA
. 
Lymphokine-activated killer cell phenomenon. Lysis of natural killer-resistant fresh solid tumor cells by interleukin 2-activated autologous human peripheral blood lymphocytes
.
J Exp Med
1982
;
155
:
1823
41
.
27.
Whiteside
TL
,
Miescher
S
,
Hurlimann
J
,
Moretta
L
,
von Fliedner
V
. 
Separation, phenotyping and limiting dilution analysis of T-lymphocytes infiltrating human solid tumors
.
Int J Cancer
1986
;
37
:
803
11
.
28.
Muul
LM
,
Spiess
PJ
,
Director
EP
,
Rosenberg
SA
. 
Identification of specific cytolytic immune responses against autologous tumor in humans bearing malignant melanoma
.
J Immunol
1987
;
138
:
989
95
.
29.
Karimi
M
,
Cao
TM
,
Baker
JA
,
Verneris
MR
,
Soares
L
,
Negrin
RS
. 
Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+ T cells and NK cells
.
J Immunol
2005
;
175
:
7819
28
.
30.
Verneris
MR
,
Karami
M
,
Baker
J
,
Jayaswal
A
,
Negrin
RS
. 
Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8+ T cells
.
Blood
2004
;
103
:
3065
72
.
31.
Nishimura
R
,
Baker
J
,
Beilhack
A
,
Zeiser
R
,
Olson
JA
,
Sega
EI
, et al
In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity
.
Blood
2008
;
112
:
2563
74
.
32.
Thorne
SH
,
Negrin
RS
,
Contag
CH
. 
Synergistic antitumor effects of immune cell-viral biotherapy
.
Science
2006
;
311
:
1780
4
.
33.
Takayama
T
,
Sekine
T
,
Makuuchi
M
,
Yamasaki
S
,
Kosuge
T
,
Yamamoto
J
, et al
Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial
.
Lancet
2000
;
356
:
802
7
.
34.
Heng
DY
,
Xie
W
,
Regan
MM
,
Warren
MA
,
Golshayan
AR
,
Sahi
C
, et al
Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study
.
J Clin Oncol
2009
;
27
:
5794
9
.
35.
Kantoff
PW
,
Schuetz
TJ
,
Blumenstein
BA
,
Glode
LM
,
Bilhartz
DL
,
Wyand
M
, et al
Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer
.
J Clin Oncol
2010
;
28
:
1099
105
.
36.
Hoos
A
,
Eggermont
AM
,
Janetzki
S
,
Hodi
FS
,
Ibrahim
R
,
Anderson
A
, et al
Improved endpoints for cancer immunotherapy trials
.
J Natl Cancer Inst
2010
;
102
:
1388
97
.