Introduction
Colorectal carcinoma (CRC) is one of the most common malignant tumors of the digestive tract and a relevant cause of cancer-related deaths. It is the third most-common tumor disease in both sexes worldwide and the second-leading cause of death among all cancers [
[1]- Li P.
- Xiao Z.
- Braciak T.A.
- Ou Q.
- Chen G.
- Oduncu F.S.
A relationship to survival is seen by combining the factors of mismatch repair status, tumor location and age of onset in colorectal cancer patients.
,
[2]- Mármol I.
- Sánchez-de-Diego C.
- Pradilla Dieste A.
- Cerrada E.
- Rodriguez Yoldi M.J.
Colorectal carcinoma: a general overview and future perspectives in colorectal cancer.
]. Over the decades, classical treatment of these tumors, i.e., surgical excision, standard chemotherapy and radiation, has been refined, resulting in significantly prolonged survival, especially for tumors detected in early stages of disease. Despite screening programs and preventive strategies, approximately 25% of patients still present with late tumor stages. Advances in surgery and expansion of systemic treatment options achieve only low improvement of median survival in patients with metastatic CRC. In recent years, mortality decreased slightly in older patients (>50 years) but remained stable in younger patients (20–49 years) [
[3]- Rawla P.
- Sunkara T.
- Barsouk A.
Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors.
,
[4]Epidemiology and mechanisms of the increasing incidence of colon and rectal cancers in young adults.
].
There is a need for effective therapeutic strategies, especially in chemorefractory tumors. CRCs with high mutational burden respond to immunotherapy. Immune checkpoint inhibitors (programmed death-1–specific antibodies as pembrolizumab or nivolumab) gain durable responses in mismatch repair-deficient-microsatellite instability-high (dMMR-MSI-H) CRC [
[5]- Le D.T.
- Uram J.N.
- Wang H.
- Bartlett B.R.
- Kemberling H.
- Eyring A.D.
- et al.
PD-1 blockade in tumors with mismatch-repair deficiency.
]. Only tumors with dMMR-MSI-H respond to immunotherapy with checkpoint inhibitors [
[5]- Le D.T.
- Uram J.N.
- Wang H.
- Bartlett B.R.
- Kemberling H.
- Eyring A.D.
- et al.
PD-1 blockade in tumors with mismatch-repair deficiency.
]. However, dMMR MSI-H tumors are found in only 15% of patients with CRC [
[6]- Guinney J.
- Dienstmann R.
- Wang X.
- de Reyniès A.
- Schlicker A.
- Soneson C.
- et al.
The consensus molecular subtypes of colorectal cancer.
,
[7]- Raut C.P.
- Pawlik T.M.
- Rodriguez-Bigas M.A.
Clinicopathologic features in colorectal cancer patients with microsatellite instability.
].
Recently, adoptive cell transfer (ACT) using tumor-infiltrating T lymphocytes (TILs), chimeric antigen receptor insertion, or T-cell receptor modification has emerged as a promising approach for tumor therapy. ACT can be used to harness the host T-cell immune response for immunotherapy in which
ex vivo mass-expanded activated TILs from resected individual tumors are reinfused to the patient. ACT using TILs achieved durable complete remission in 20%–25% of patients with metastatic melanoma [
[8]- Rosenberg S.A.
- Yang J.C.
- Sherry R.M.
- Kammula U.S.
- Hughes M.S.
- Phan G.Q.
- et al.
Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy.
]. Successful treatments also have been reported for other solid tumors, such as cervical cancer and ovarian cancer [
[9]- Fujita K.
- Ikarashi H.
- Takakuwa K.
- Kodama S.
- Tokunaga A.
- Takahashi T.
- et al.
Prolonged disease-free period in patients with advanced epithelial ovarian cancer after adoptive transfer of tumor-infiltrating lymphocytes.
,
[10]- Stevanović S.
- Draper L.M.
- Langhan M.M.
- Campbell T.E.
- Kwong M.L.
- Wunderlich J.R.
- et al.
Complete regression of metastatic cervical cancer after treatment with human papillomavirus-targeted tumor-infiltrating T cells.
].
Previous studies showed that interactions among the immune microenvironment and the mediated immune response are related to a favorable prognosis in CRC [
11- Mlecnik B.
- Bindea G.
- Angell H.K.
- Sasso M.S.
- Obenauf A.C.
- Fredriksen T.
- et al.
Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients.
,
12- Mlecnik B.
- Tosolini M.
- Kirilovsky A.
- Berger A.
- Bindea G.
- Meatchi T.
- et al.
Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction.
,
13- Pagès F.
- Berger A.
- Camus M.
- Sanchez-Cabo F.
- Costes A.
- Molidor R.
- et al.
Effector memory T cells, early metastasis, and survival in colorectal cancer.
,
14- Pagès F.
- Kirilovsky A.
- Mlecnik B.
- Asslaber M.
- Tosolini M.
- Bindea G.
- et al.
In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer.
]. A high degree of tumor-infiltrating lymphocytes (TILs) in specimens is associated with decreased tumor invasiveness, less nodal involvement and improvement in survival [
15- Berntsson J.
- Nodin B.
- Eberhard J.
- Micke P.
- Jirström K.
Prognostic impact of tumour-infiltrating B cells and plasma cells in colorectal cancer.
,
16- Däster S.
- Eppenberger-Castori S.
- Hirt C.
- Zlobec I.
- Delko T.
- Nebiker C.A.
- et al.
High frequency of CD8 positive lymphocyte infiltration correlates with lack of lymph node involvement in early rectal cancer.
,
17- Governa V.
- Trella E.
- Mele V.
- Tornillo L.
- Amicarella F.
- Cremonesi E.
- et al.
The interplay between neutrophils and CD8(+) T cells improves survival in human colorectal cancer.
,
18The PD-1 expressing immune phenotype of T cell exhaustion is prominent in the 'immunoreactive' microenvironment of colorectal carcinoma.
,
19- Salama P.
- Phillips M.
- Grieu F.
- Morris M.
- Zeps N.
- Joseph D.
- et al.
Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer.
].
Despite the enormous disease burden and large number of premature deaths caused by CRC, and despite the good rationale for an immunotherapeutic approach, this entity has been only a minor object in cell therapy research [
20- Hom S.S.
- Rosenberg S.A.
- Topalian S.L.
Specific immune recognition of autologous tumor by lymphocytes infiltrating colon carcinomas: analysis by cytokine secretion.
,
21- Tran E.
- Robbins P.F.
- Lu Y.C.
- Prickett T.D.
- Gartner J.J.
- Jia L.
- et al.
T-cell transfer therapy targeting mutant KRAS in cancer.
,
22- Vose B.M.
- Gallagher P.
- Moore M.
- Schofield P.F.
Specific and non-specific lymphocyte cytotoxicity in colon carcinoma.
]. Moreover, studies on ACT often lack controlled and comparable expansion processes as well as selected ACT-relevant T-cell populations.
The aim of this study was to investigate the feasibility of isolating TILs from CRC tissue samples and mass expanding of TILs for ACT in a controlled two-phase process in a perfusion bioreactor. The expanded TILs were characterized according to phenotype, and activity was determined based on cytotoxicity to different stimulations.
Methods
Tumor samples
Tumor tissue samples were obtained from patients with primary CRC undergoing surgical resection without preoperative chemotherapy, radiotherapy or immunotherapy at the Department of General, Visceral, Thoracic, and Vascular Surgery, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, as part of a study to analyze essential fatty acids/lipid mediators and their function in solid tumors (Ethics Committee of the Brandenburg Medical School No. Z-03-20170508). All patients gave their written informed consent to participate in the study, and included were 12 primary colon tumors obtained by surgical resection with curative intent. Additional clinical data of patients are listed in
Table 1. After surgical resection, tumors were localized in the specimen and opened by an incision from the luminal or extraluminal side. The tumor center, invasive margin, and peritumoral environment were identified, and tissue samples were collected from the invasive margin area.
Table 1Patients and tumor characteristics.
f, female; m, male; MSI-H/MSS, microsatellite instability or stability; UICC, Union for International Cancer Control.
Pathological characterization and transportation to in vitro processing
TILs were quantified in the obtained tissue samples before expansion and their phenotype was characterized. For this process, paraffin-embedded tissue samples fixed in formalin were used and hematoxylin-eosin (H&E)-stained slides were prepared.
The standardized method for the assessment of TILs in solid tumors on H&E sections was applied according to the guidelines of the International Immuno-Oncology Biomarker Working Group [
[23]- Hendry S.
- Salgado R.
- Gevaert T.
- Russell P.A.
- John T.
- Thapa B.
- et al.
Assessing tumor-infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the international immunooncology biomarkers working group: part 1: assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research.
]. Semiquantitative H&E evaluation of the TILs was performed at 200–400× magnification. In addition, tissue samples were examined by immunohistochemistry for infiltration with CD4- and CD8-positive T cells (
supplementary Figure 1).
The following antibodies were used for immunostaining: monoclonal mouse anti-human CD4 (Agilent Technologies [Santa Clara, CA, USA], 4B12, ready-to-use) and monoclonal mouse anti-human CD8 (Agilent Technologies, C8/144B, ready-to-use).
The immuno-stained slides were analyzed semi-quantitatively by a pathologist, who assessed the ratio between the number of positively stained cells and the total number of cells in five high power fields. Areas with significant necrosis, hemorrhage, as well as histologic artifacts, were excluded from analysis.
Tumor tissue samples from each donor were transported to the laboratory under antibiotic-antifungal conditions (CellGenix GMP DC Medium [Sartorius CellGenix, Freiburg, Germany], 10% human serum [PAN Biotech, Bavaria, Germany], 100 units penicillin, 0.10 mg streptomycin and 0.25 μg amphotericin B per mL [Sigma–Aldrich, St. Louis, MO, USA]).
As a result of contamination events during primary cultivation and the loss of two tumor samples, the transportation medium was stowed with double-concentrated antibiotic-antimycotic reagent (200 units penicillin, 0.20 mg streptomycin and 0.50 μg amphotericin B per milliliter).
Activation and expansion of TIL in perfusion bioreactor mode
Preparation of tumor tissue, cultivation and
ex vivo expansion of tumor infiltrating lymphocytes was done in a proprietary cultivation process developed by Zellwerk GmbH [
[24]Hoffmeister, H. (2019). Meander bioreactor and method for the isolation and proliferation of cells from parts from tumors, metastases and other tissues. Germany patent application.
,
[25]Hoffmeister, H. (2020). Meandering perfusion bioreactor for the differentiation, activation, stimulation and separation of cells. Germany patent application.
]. Tumor tissue material was sliced into 2 × 2 × 2-mm pieces and filled into 15 mL of complete cultivation medium (CCM), composed of CellGenix GMP DC Medium (Sartorius CellGenix), 10% human serum (PAN Biotech), 1% antibiotic antimycotic solution (100 units penicillin, 0.10 mg streptomycin and 0.25 μg amphotericin B per milliliter) (Sigma-Aldrich) and 1000 IU/mL human IL-2 (Proleukin; Clinigen, Burton upon Trent, UK). After adding 10 ng per milliliter of human IL-12 (premium grade; Miltenyi Biotec, Bergisch Gladbach, Germany), the suspension was transferred via port directly into a perfusion bioreactor. Outgrowth, activation and expansion of TIL were carried out in a single use, GMP-compliant and completely closed perfusion bioreactor 30MM. The bioreactor runs were operated on a platform of GMP breeder, Control Unit (both from Zellwerk GmbH, Oberkrämer, Germany) and associated software [
[26]- Bröker K.
- Sinelnikov E.
- Gustavus D.
- Schumacher U.
- Pörtner R.
- Hoffmeister H.
- et al.
Mass production of highly active NK Cells for cancer immunotherapy in a GMP conform perfusion bioreactor.
,
[27]- Pörtner R.
- Sebald C.
- Parida S.
- Hoffmeister H.
Single-use bioreactors for manufacturing of immune cell therapeutics.
]. This platform allows measurement and control of medium temperature, pH and pO
2 (set point for temperature: 36.5°C; for pH: 7.2–7.3 [regulated by CO
2 influx]; for pO
2: 25% [using a gas mixture of 25% O
2 and 75% N
2]).
The first cultivation period within a bioreactor run comprised 6–8 days. Inoculation was performed with different numbers of tissue pieces in CCM with 10 ng per milliliter human IL-12 under static aeration conditions (5% CO2, 25% O2, 70% N2). In the first two experiments, we inoculated 50 tissue pieces, each in a total volume of 400 mm3. Later, total tissue volume was reduced to 240 mm3 (30 pieces, four donors) and subsequently, from each of 5 donors 10 pieces (80 mm3) were inoculated, leading to the best yield. At days 3–5 within the first cultivation period, outgrowth and expansion of TIL is initiated by a short activation burst: A cocktail containing 30 ng per milliliter anti-human CD3 antibody (clone OKT3; Miltenyi Biotec) and a number of 2 million allogenic human feeder cells per tumor piece (irradiated with 55 Gy), is infused once into a bioreactor. During the next 3–5 days, an accelerated outgrowth of TILs is observed.
The second cultivation period begins with starting medium circulation in the bioreactor vessel as well as a continuous influx of fresh CCM. The culture process is from this time automatically controlled by an algorithm based on continuous measurements of pH, pO2 and temperature in the circulating medium and provides upgrowing cells with fresh medium.
As CCM is only supplemented with interleukin (IL)-2, the activating substance IL-12 is washed out after approximately 1 day of fresh CCM supply. Medium circulation and needed fresh medium influx over the whole expansion phase of TIL until harvesting is steered automatically, guaranteeing homogenous nutrient supply. To achieve a rolling movement, changing the touching areas between the sedimented TIL, the bioreactor vessel needs a short gently shaking every 5–7 days.
Over the whole culturing time, supernatant samples were regularly taken and concentrations of glucose and lactate were routinely measured using a biochemical analyzer (YSI 2700 Select, YSI).
When the lactate concentration reached a target value of 1 g/L, influx of fresh medium (perfusion rate) was adjusted manually to maintain this value. Cells were expanded until an average final daily lactate production of 80–120 mg per day was reached. The cell number reached was approximately 1.5 to 2.5 × 109 within a period of 3–4 weeks. The bioreactor was shaken to suspend the cells before taking samples for cell counting. Cells were harvested, contemporaneous separated from tissue material by transferring them by gravity over the mesh port, cryopreserved and stored in liquid nitrogen until used for the second expansion phase.
For the second expansion phase, cells were thawed, washed and mixed with 20 mL of complete cultivation medium (as mentioned previously) and filled directly into the bioreactor. The bioreactor was inoculated with approximately 300 × 10
6 cells. After 3 hours, when cells had settled down, medium circulation and aeration were enabled. During this expansion, the observed process parameters were comparable to those of the first expansion phase. The tumor volume or inoculated cells used for the first or second expansion phase, the duration of cultivation and the yield of harvested cells are listed in
Table 2.
Table 2Cultivation parameters: Starting material, cell yield, duration of cultivation, and metabolites in bioreactor in first and second expansion phases.
Phenotypic characterization by flow cytometry
The phenotype of expanded cells of each bioreactor cultivation was analyzed by using a BD Accuri C6 Plus (BD Biosciences, Franklin Lakes, NJ, USA) or MACSQuant Analyzer 10 flow cytometer (Miltenyi Biotec). Characterization of different lineage markers and other expressed surface markers was performed directly after bioreactor harvesting as well as after short-term cultivation of cryopreserved cells.
For analysis, cells were washed, followed by staining with different sets of antibodies. Anti-CD3 PerCP-Cy5.5 (clone: UCHT1; BioLegend, San Diego, CA, USA), anti-CD4 FITC (clone: RPA-T4; BioLegend), anti-CD8 FITC or PE (clone: SK1; BioLegend), anti-CD28 APC (clone: CD28.2; BioLegend); anti-CCR7 APC (clone: G043H7; BioLegend), anti-CD25 APC (clone: M-A251; BD Biosciences); anti-CD27 PE (clone: M-T271; BD Biosciences); anti-CD45RO PE (clone: UCHL1; BD Biosciences); anti-CD56 FITC (clone: B159; BD Biosciences); anti-CD127 PE (clone: HIL-7R-M21; BD Biosciences); anti-CD183 PE (clone: 1C6/CXCR3; BD Biosciences); anti-CD196 APC (clone: 11A9; BD Biosciences) (panel 1) or Anti-CD3 PE-Vio615 (clone: REA613), anti-CD4 APC-Vio770 (clone: REA623), anti-CD8 VioGreen (clone: REA734), anti-CD25 PE (clone: REA945); anti-CD27 PE (REA499); anti-CD28 APC (REA612); anti-CD45RO FITC (clone: REA611); anti-CD56 APC (clone: REA196); anti-CD127 APC (clone: REA614); anti-CD183 APC (REA232); anti-CD196 PE-Vio770 (REA190); anti-CCR7 PE-Vio770 (clone: REA546) (panel 2, all Miltenyi Biotec) were used as lineage markers for T cells (Th, Tc, Treg) and NK cells, and combined with either T cell subtype markers for naïve, central memory (TCM), effector or effector memory T cells (Teff, TEM) and for T helper cells (Th1, Th2, Th17). Cells were stained for 30 min at room temperature, washed and finally re-suspended in fresh staining buffer.
Cell culture
Tumor-infiltrating lymphocytes
Before any analytical measurements, cryopreserved TILs were re-vitalized in short-term culture. The cells were thawed, followed by washing (350g, 5 min) with pre-warmed cultivation medium. Cells were then resuspended in CCM, transferred into flasks and cultivated for 48 h at 37°C, 25% O2, 5.5% CO2. Afterwards, cells were collected, centrifuged (350g, 5 min) and resuspended in TIL-CCM.
CRC cell lines
For co-cultivation of TIL and CRC cell lines, cryopreserved aliquots of each cell line (COLO678 [ACC194], HCT116 [ACC581], JVE127 [ACC813]; DSMZ) were thawed and then mixed with prewarmed cultivation medium (CRC-M; RPMI-1640 [Gibco/Thermo Fisher Scientific, Waltham, MA, USA], 10% fetal bovine serum [Sigma-Aldrich], 1% antibiotic antimycotic solution (100 units penicillin, 0.10 mg streptomycin and 0.25 μg amphotericin B per milliliter) [Sigma Aldrich]). Cells were then resuspended in CRC-M, transferred to flasks and cultured at 37°C and 5.5% CO2.
Functional assays
Qualitative interferon (IFN)γ staining assay
The potential of TILs to express IFNγ was analyzed using a MACSQuant Analyzer 10 flow cytometer (Miltenyi Biotec). To assess the intracellular IFNγ expression of TILs, cells were cultivated as described previously. After short-term cultivation, cells were resuspended in complete supplemented medium (included 10% human serum, 1% antibiotic antimycotic solution, IL-2) in a concentration of 2 million cells per milliliter. Then, 50 µL of this suspension were transferred into 96-well round-bottom plates. Cells were stimulated with anti-CD3 antibody (clone: OKT3, Miltenyi Biotec) at a final concentration of 30 ng per milliliter. Phorbol-12-myristat-13-acetat (PMA) was used as an alternative stimulation reagent and as a positive control (final conc. 2.5 µg/mL). After 1 h of incubation at 37°C, 25% O2, 5.5% CO2, 50 µL of GolgiStop (1:150, BD Biosciences) and GolgiPlug (1:100, BD Biosciences), diluted in complete supplemented medium (included 10% human serum, 1% antibiotic antimycotic solution, IL-2, anti-CD3 Ab), were added. After further 5 h of incubation, cells were collected, centrifuged and incubated together with TruStain FcX (BioLegend) for 10 min at room temperature. Afterwards, surface markers were stained with anti-CD3 PerCP-Cy5.5, anti-CD4 FITC and anti-CD8 PE antibodies for 30 min at room temperature (Ab details described previously: panel 1). The fixation/permeabilization (BD Cytofix/Cytoperm fixation/permeabilization kit; BD Biosciences) overnight at 4°C was performed after washing with staining buffer (350g, 5min). At the next day, cells were washed with perm/wash solution and stained with anti-IFNγ APC (clone: 4S.B3, BioLegend) antibody for 30 min at room temperature. After final washing, cells were resuspended in staining buffer and stored at 4°C until measurement.
Quantitative cytokine assay
The concentrations of secreted cytokines were analyzed by using a MACSQuant Analyzer 10 flow cytometer (Miltenyi Biotec). To assess the potential of TILs to secret different cytokines, cells were cultivated as described previously. After short-term cultivation, cells were re-suspended in complete supplemented medium (included 10% human serum, 1% antibiotic antimycotic solution, IL-2) in a concentration of 1 million cells per milliliter. Then, 200 µL of this suspension were transferred into 96-well round-bottom plates. Cells were stimulated with anti-CD3 Ab at a final concentration of 30 ng/mL (clone: OKT3, Miltenyi Biotec). Further, after 6 h of incubation, supernatants were collected, centrifuged (10 000g, 1 min) and the cell-free supernatants were frozen at –20°C.
To measure different cytokines in parallel, Miltenyi Biotec MACSPlex cytotoxic assays were used. In this assay, we combined the following products, MACSPlex mix cytotoxic basic kit, MACSPlex mix cytotoxic standard and MACSPlex mix cytotoxic reagents kits for IFNγ and tumor necrosis factor (TNF)α.
At the day of measurement, supernatants were thawed, centrifuged (10 000g, 10 min) and prepared as described by the manufacturer. To analyze and quantify, amounts of cytokines were measured by MACSQuant Analyzer Express Mode for MACSQuant Instrument.
Degranulation and intracellular cytokine assay
Degranulation and intracellular cytokine expression were analyzed using a MACSQuant Analyzer 10 flow cytometer (Miltenyi Biotec). To investigate the degranulation and intracellular cytokine expression (IFNγ, TNFα) of TILs induced by tumor-cell-specific stimuli, three CRC-cell lines were co-cultivated with TILs. Two to three days before co-cultivation, CRC cell lines were resuspended in CRC-M (RPMI-1640, 10% fetal bovine, 1% antibiotic antimycotic solution) at a concentration of half a million cells per milliliter. Then, 100 µL of this suspension was transferred into 96-well flat-bottom plate and cultivated at 37°C, 5.5% CO2. On the day of co-cultivation, short-term TILs (as described previously) were resuspended in complete supplemented medium (containing 10% human serum, 1% antibiotic antimycotic solution and IL-2) at a concentration of 10 million cells per milliliter. Then, 50 µL of this suspension was transferred into 96-well flat-bottom plates containing CRC-cell lines (after removal of CRC medium). CD107a-Ab (anti-CD107a APC [REA792]; Miltenyi Biotec) and/or PMA (duplicates for each CRC-cell line as stimulation control; final concentration 2.5 µg/mL) were transferred into TIL-CRC-cocultures. After 1 h of incubation at 37°C, 25% O2, 5.5% CO2, 20 µL of GolgiStop (1:132, BD Biosciences) and GolgiPlug (1:99, BD Biosciences), diluted in complete supplemented medium, were added. After further 5 h of incubation, cells were collected, centrifuged and washed with staining buffer (autoMACS Rinsing solution incl. BSA; Miltenyi Biotec). Cells were then fixed and stained as described by the manufacturer (extracellular: anti-CD3 PE-Vio770 [REA613], anti-CD4 APC-Vio770 [REA623], anti-CD8 VioGreen [REA734]; intracellular: anti-IFNγ FITC [REA600] and anti-TNFα PE [REA656]; all antibodies from Miltenyi Biotec). After the final wash, cells were resuspended in staining buffer and immediately measured. Each TIL-CRC co-culture was analyzed in triplicate.
Statistical analyses
Statistical analyses were performed using GraphPad Prism 9 (GraphPad Software, Inc., San Diego, CA, USA). Statistical differences between unstimulated and stimulated samples were determined using a nonparametric one-way analysis of variance rank test (Friedmann) with Dunn multiple comparisons test.
Discussion
In the development of new therapeutic strategies for colorectal cancer, it was found that only the less-frequent tumors with dMMR-MSI-H respond to immunotherapy with checkpoint inhibitors [
[5]- Le D.T.
- Uram J.N.
- Wang H.
- Bartlett B.R.
- Kemberling H.
- Eyring A.D.
- et al.
PD-1 blockade in tumors with mismatch-repair deficiency.
]. Because a high density of tumor-infiltrating lymphocytes is associated with a better prognosis in CRC [
11- Mlecnik B.
- Bindea G.
- Angell H.K.
- Sasso M.S.
- Obenauf A.C.
- Fredriksen T.
- et al.
Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients.
,
12- Mlecnik B.
- Tosolini M.
- Kirilovsky A.
- Berger A.
- Bindea G.
- Meatchi T.
- et al.
Histopathologic-based prognostic factors of colorectal cancers are associated with the state of the local immune reaction.
,
13- Pagès F.
- Berger A.
- Camus M.
- Sanchez-Cabo F.
- Costes A.
- Molidor R.
- et al.
Effector memory T cells, early metastasis, and survival in colorectal cancer.
,
14- Pagès F.
- Kirilovsky A.
- Mlecnik B.
- Asslaber M.
- Tosolini M.
- Bindea G.
- et al.
In situ cytotoxic and memory T cells predict outcome in patients with early-stage colorectal cancer.
], ACT using TILs is a reasonable approach that takes advantage of the patient's specific T-cell immune response towards the tumor.
The potential success of ACT depends on both the quantity and quality of expanded TILs from tumor tissue. In terms of quantity, a number of 1 × 10
10 expanded TILs is considered an appropriate dose for a therapeutic infusion [
[28]Pörtner, R., Parida, S., Schaffer, C., and Hoffmeister, H. (2018). "Landscape of manufacturing process of ATMP cell therapy products for unmet clinical needs." 2018 May 2; Available from: http://dx.doi.org/10.5772/intechopen.69335.
,
[29]- Weber J.
- Atkins M.
- Hwu P.
- Radvanyi L.
- Sznol M.
- Yee C
White paper on adoptive cell therapy for cancer with tumor-infiltrating lymphocytes: a report of the CTEP subcommittee on adoptive cell therapy.
]. However, in a pilot study using expanded T cells from CRC sentinel lymph nodes for ACT, complete tumor responses were achieved with only 8 × 10
7 cells in metastatic CRC [
[30]- Karlsson M.
- Marits P.
- Dahl K.
- Dagöö T.
- Enerbäck S.
- Thörn M.
- et al.
Pilot study of sentinel-node-based adoptive immunotherapy in advanced colorectal cancer.
]. It must be taken into account that the density of TILs is lower in gastrointestinal (GI) tumors compared with, for example, melanoma [
[31]- Turcotte S.
- Gros A.
- Hogan K.
- Tran E.
- Hinrichs C.S.
- Wunderlich J.R.
- et al.
Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy.
].
In our study, the obtained tumor tissue samples were subjected to pathological screening, in which TILs were examined in H&E and by immunostaining. Pathological screening revealed TIL infiltration rates of 5%–60% and a varying CD4+/CD8+ ratio in favor of CD4+ cells (mean 62/30%). Regardless of these initial infiltration rates, our data show that expansion of TILs in a low-scaled, two-phase process in the Zellwerk ZRP bioreactor from surgically resected primary CRC resulted in a relevant number of approximately 2 × 109 cells. There was no correlation between TIL infiltration rates on pathological screening and the number of expanded cells. The second phase can be scaled up to manufacture more than 1010 TILs, using a bioreactor with 5 times more expansion area. TIL expansion was performed in a two-phase process to separate cells from medium and residual tumor tissue at the end of phase 1 to avoid tumor-related inhibitory effects.
We observed a strong dependency of TIL outgrowth on tumor mass during starting period. Starting expansion with a relatively large tumor volume (400 mm3; 50 pieces), we noticed limited cell yield after phase 1. Reducing tumor volume (to 240 mm3 and later to 80 mm3) significantly increased cell yield after phase 1 and resulted in an ACT-relevant number of TILs after phase 2. Therefore, the quantity of 80 mm3 was set as an optimized standard. The reason for the phenomenon described is not clear, but is probably related to tumor-induced immune-inhibitory effects with higher volumes of tumor tissue present.
TILs were manufactured on a platform on which perfusion type bioreactors were operated in an automatically controlled culturing process. The outgrowth of TILs from tumor pieces, activation and long-term expansion was performed in a closed perfusion bioreactor system developed at Zellwerk GmbH and established for many years [
[26]- Bröker K.
- Sinelnikov E.
- Gustavus D.
- Schumacher U.
- Pörtner R.
- Hoffmeister H.
- et al.
Mass production of highly active NK Cells for cancer immunotherapy in a GMP conform perfusion bioreactor.
,
[27]- Pörtner R.
- Sebald C.
- Parida S.
- Hoffmeister H.
Single-use bioreactors for manufacturing of immune cell therapeutics.
,
[28]Pörtner, R., Parida, S., Schaffer, C., and Hoffmeister, H. (2018). "Landscape of manufacturing process of ATMP cell therapy products for unmet clinical needs." 2018 May 2; Available from: http://dx.doi.org/10.5772/intechopen.69335.
]. This meander bioreactor is characterized by a directional laminar flow of the medium, which allows homogeneous distribution of nutrients and gases over the cultivation period and minimizes disruption of cell–cell and cell–surface contact.
Regarding the subtypes of expanded TILs, CD8
+ T cells are the most important cell population for the anti-tumor response [
[32]- Dudley M.E.
- Gross C.A.
- Langhan M.M.
- Garcia M.R.
- Sherry R.M.
- Yang J.C.
- et al.
CD8+ enriched "young" tumor infiltrating lymphocytes can mediate regression of metastatic melanoma.
,
[33]- Prieto P.A.
- Durflinger K.H.
- Wunderlich J.R.
- Rosenberg S.A.
- Dudley M.E.
Enrichment of CD8+ cells from melanoma tumor-infiltrating lymphocyte cultures reveals tumor reactivity for use in adoptive cell therapy.
]. It was postulated that tumor-specific CD4
+ T cells in addition to CD8
+ T cells might enhance and prolong the anti-tumor response in ACT [
[34]- Wang L.X.
- Shu S.
- Disis M.L.
- Plautz G.E.
Adoptive transfer of tumor-primed, in vitro-activated, CD4+ T effector cells (TEs) combined with CD8+ TEs provides intratumoral TE proliferation and synergistic antitumor response.
].
In our study, the CD4
+/CD8
+ ratio constantly shifted towards CD8
+ cells during TIL expansion, resulting in about 65% after phase 1 and about 86% CD8
+ cells after phase 2. This is in contrast to other studies, which reported that a lower proportion of CD8
+ cells were expanded from GI tumors compared to melanoma, in which >70% CD8+ cells are routinely found [
[20]- Hom S.S.
- Rosenberg S.A.
- Topalian S.L.
Specific immune recognition of autologous tumor by lymphocytes infiltrating colon carcinomas: analysis by cytokine secretion.
,
[31]- Turcotte S.
- Gros A.
- Hogan K.
- Tran E.
- Hinrichs C.S.
- Wunderlich J.R.
- et al.
Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy.
]. However, in these studies, TILs were mainly expanded from metastases of GI tumors. It remains unclear whether the different tumor material or the expansion process account for the different results.
In this study, double-positive (CD4
+ CD8
+) cells were found in a small proportion of approximately 2.4% of TILs after expansion phase 1 and 0.8% after phase 2, respectively. Tumor reactivity was noted for these double-positive TILs in CRC. However, because the frequency of double-positive TILs is increased in metastatic CRC, it has been postulated that this subset of TILs may play a role in the metastatic process by downregulating the immune response to the tumor [
[35]- Sarrabayrouse G.
- Corvaisier M.
- Ouisse L.H.
- Bossard C.
- Le Mével B.
- Potiron L.
- et al.
Tumor-reactive CD4+ CD8αβ+ CD103+ αβT cells: a prevalent tumor-reactive T-cell subset in metastatic colorectal cancers.
]. The significance of the double-positive cells among expanded TILs is not yet determined.
Further characterization of ACT-relevant TIL-subtypes includes markers for antigen experience and differentiation of effector (T
eff) and regulatory T cells (T
reg). Previous preclinical and clinical studies of ACT have mostly used unselected T cells, including cytotoxic, helper, and even immunosuppressive T
reg cells [
[30]- Karlsson M.
- Marits P.
- Dahl K.
- Dagöö T.
- Enerbäck S.
- Thörn M.
- et al.
Pilot study of sentinel-node-based adoptive immunotherapy in advanced colorectal cancer.
,
[36]- Zhen Y.H.
- Liu X.H.
- Yang Y.
- Li B.
- Tang J.L.
- Zeng Q.X.
- et al.
Phase I/II study of adjuvant immunotherapy with sentinel lymph node T lymphocytes in patients with colorectal cancer.
]. More precise characterization and selection of the T cells used could be a promising approach to improve efficacy and safety of ACT.
It has been shown that a high T
eff/T
reg ratio is essential for effective immunosurveillance of GI tumors and is therefore target for an ACT-relevant TIL population [
[37]- Lutz E.R.
- Wu A.A.
- Bigelow E.
- Sharma R.
- Mo G.
- Soares K.
- et al.
Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation.
,
[38]- Niccolai E.
- Ricci F.
- Russo E.
- Nannini G.
- Emmi G.
- Taddei A.
- et al.
The different functional distribution of "not effector" T cells (Treg/Tnull) in colorectal cancer.
]. The T cells found in the inhibitory tumor microenvironment are predominantly Treg cells, but appear to have a plastic phenotype that can be polarized toward effector function upon expansion in IL-2 [
[31]- Turcotte S.
- Gros A.
- Hogan K.
- Tran E.
- Hinrichs C.S.
- Wunderlich J.R.
- et al.
Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy.
,
[37]- Lutz E.R.
- Wu A.A.
- Bigelow E.
- Sharma R.
- Mo G.
- Soares K.
- et al.
Immunotherapy converts nonimmunogenic pancreatic tumors into immunogenic foci of immune regulation.
,
[39]- Quezada S.A.
- Peggs K.S.
- Curran M.A.
- Allison J.P
CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells.
].
CD45RO has been shown to be a suitable marker for (tumor antigen)-activated CD8
+ T effector cells, which have the potential to elicit an anti-tumor immune response [
[40]Tumor-infiltrating CD45RO(+) Memory T lymphocytes predict favorable clinical outcome in solid tumors.
,
[41]- Michie C.A.
- McLean A.
- Alcock C.
- Beverley P.C.
Lifespan of human lymphocyte subsets defined by CD45 isoforms.
]. Moreover, CD45RO
+ T cells are associated with improved survival [
[40]Tumor-infiltrating CD45RO(+) Memory T lymphocytes predict favorable clinical outcome in solid tumors.
]. Therefore, aiming for a high proportion of CD45RO
+ T cells could increase effectiveness of TILs in the context of ACT-approaches.
CCR7 expression divides memory T cells into two functional subsets. CCR7
− memory cells have direct effector function and are therefore considered effector memory T cells (T
EM). CCR7
+ cells are central memory T cells (T
CM) that express lymph node homing receptors but have no immediate effector function [
[42]- Sallusto F.
- Lenig D.
- Förster R.
- Lipp M.
- Lanzavecchia A.
Two subsets of memory T lymphocytes with distinct homing potentials and effector functions.
].
In the current study, the phenotype of CD8+ effector memory cells (CD45RO+/CCR7−) accounted for approximately 60% of TILs after expansion phase 1 and 73% of TILs after phase 2, respectively. This suggests that this cell population could have a long-term cytotoxic effect against tumor cells.
To investigate the functional potential of the obtained TILs upon nonspecific stimulation, we chose a time frame of 6 h of stimulation suitable for testing also in the context of further clinical use, both with regard to quantification of intracellular cytokine expression as well as for quantification of degranulation in the supernatant.
Anti-CD3 stimulation and intracellular cytokine staining revealed CD8
+ IFNγ
+ cells in 3% of TILs in expansion phase 1 and 4.2% of TILs in phase 2, respectively. These data appear to be small compared with reports in the literature. It should be noted, however, that the duration of stimulation in these studies usually ranged from 12 hours up to several days [
[34]- Wang L.X.
- Shu S.
- Disis M.L.
- Plautz G.E.
Adoptive transfer of tumor-primed, in vitro-activated, CD4+ T effector cells (TEs) combined with CD8+ TEs provides intratumoral TE proliferation and synergistic antitumor response.
]. PMA stimulation lasting 6 hours resulted in CD8
+ IFNγ
+ cells in 50% of TILs harvested in phase 1 and in 55% of TILs in expansion phase 2.
In addition, quantification of degranulation showed relevant cytokine secretion in the supernatant upon both anti-CD3 and PMA stimulation for 6 hours. Overall, the functional potential of TILs expanded in our study was confirmed upon nonspecific stimulation.
In conclusion, a reliable, standardized bioreactor process for TIL expansion from primary CRC tissue samples was established, and a relevant number of TILs of an ACT-relevant subtype with functional potential could be obtained (the entire process, exemplified in
supplementary Figure 4 and
supplementary Figure 5). In this process, the Zellwerk ZRP bioreactor enabled a high degree of standardization of immune cell mass production in a closed environment under constant conditions.
The present study investigated the feasibility of expanding TIL from resected primary CRC tumors, which were predominantly mismatch repair-proficient-microsatellite instability-low (pMMR-MSI-L), and established high-volume immune cell generation also from these tumors, which are not sensitive to checkpoint inhibitor immunotherapy approaches. Therefore, ACT options using TILs could be an approach to harness an anti-tumor immune response for this subset of CRC as well.
However, this study did not investigate the cytotoxic functional potential of expanded TILs when stimulated with autologous tumors cells. At the time of the investigation, autologous tumor material was no longer available. As an approximation, we examined the functional potential of TILs when challenged with a series of commercially available CRC tumor cell lines. However, degranulation and intracellular cytokine expression (IFNγ, TNFα) after short-time challenge with CRC cell lines did not demonstrate the cytotoxic potential of the expanded TILs. When co-cultured with PMA as a stimulation control, TILs were shown to have functional potential (compared to observations in assays without tumor cell lines). However, the CRC cell lines used in this study did not achieve specific stimulation of expanded TILs. It has been previously reported that GI tumors have unique mutations and antitumor T-cell responses are directed to neoantigens and cryptic peptides specific to each individual patient [
[20]- Hom S.S.
- Rosenberg S.A.
- Topalian S.L.
Specific immune recognition of autologous tumor by lymphocytes infiltrating colon carcinomas: analysis by cytokine secretion.
,
[22]- Vose B.M.
- Gallagher P.
- Moore M.
- Schofield P.F.
Specific and non-specific lymphocyte cytotoxicity in colon carcinoma.
,
[31]- Turcotte S.
- Gros A.
- Hogan K.
- Tran E.
- Hinrichs C.S.
- Wunderlich J.R.
- et al.
Phenotype and function of T cells infiltrating visceral metastases from gastrointestinal cancers and melanoma: implications for adoptive cell transfer therapy.
,
[43]- Schwarz S.
- Schmitz J.
- Löffler M.W.
- Ghosh M.
- Rammensee H.G.
- Olshvang E.
- et al.
T cells of colorectal cancer patients' stimulated by neoantigenic and cryptic peptides better recognize autologous tumor cells.
]. Therefore, commercially available colon cancer cell lines are probably not a good model for demonstrating specific cytotoxicity of expanded TILs. In further studies, we will evaluate the cytotoxic function of expanded TILs when stimulated with either cryopreserved autologous tumor cell suspensions, tumor cell-derived material or autologous cell lines.
Furthermore, it will be the next step to test TIL populations expanded from patients using the method described here in a broader range of
in vitro experiments to study a wide range of TIL function modifiers, e.g., polyunsaturated fatty acids and lipid mediators that have a role in colon tumorigenesis [
44- Gottschall H.
- Schmocker C.
- Hartmann D.
- Rohwer N.
- Rund K.
- Kutzner L.
- et al.
Aspirin alone and combined with a statin suppresses eicosanoid formation in human colon tissue.
,
45- Nowak J.
- Weylandt K.H.
- Habbel P.
- Wang J.
- Dignass A.
- Glickman J.N.
- et al.
Colitis-associated colon tumorigenesis is suppressed in transgenic mice rich in endogenous n-3 fatty acids.
,
46- Rohwer N.
- Kuhl A.A.
- Ostermann A.I.
- Hartung N.M.
- Schebb N.H.
- Zopf D.
- et al.
Effects of chronic low-dose aspirin treatment on tumor prevention in three mouse models of intestinal tumorigenesis.
,
47- Schmocker C.
- Gottschall H.
- Rund K.M.
- Kutzner L.
- Nolte F.
- Ostermann A.I.
- et al.
Oxylipin patterns in human colon adenomas.
]. In addition, factors such as preoperative immune-nutrition and gut microbiota influence the composition and function of TILs in colorectal cancer and are therefore a promising subject for further studies [
[48]- Caglayan K.
- Oner I.
- Gunerhan Y.
- Ata P.
- Koksal N.
- Ozkara S.
The impact of preoperative immunonutrition and other nutrition models on tumor infiltrative lymphocytes in colorectal cancer patients.
].
Based on these planned
in vitro cytotoxic function experiments of the expanded TILs, future
in vivo studies could be envisaged. Notably, because the complex tumor microenvironment may affect TIL function
in vivo, in vitro data can only serve as an approximation. The main obstacles to ACT
in vivo appear to be short-term persistence and poor trafficking of immune cells within the immunosuppressive tumor microenvironment, anergy and poor and insufficient proliferation of immune cells [
[49]- Fan J.
- Shang D.
- Han B.
- Song J.
- Chen H.
- Yang J.M
Adoptive cell transfer: Is it a promising immunotherapy for colorectal cancer?.
], effects that can only be evaluated and understood
in vivo.
We focused on CRC primary tumors when establishing the TIL expansion process described here in order to exclude any influences of previous therapies (radiation, chemotherapy, immunotherapy). However, given that ACT approaches might primarily be ultima ratio treatment options in heavily pretreated patients, primary tumor tissue is unlikely to be the material of choice for TIL expansion in these cases. Therefore, in a next step we will investigate whether the promising results of TIL expansion from CRC primary tumors presented here also apply to TIL expansion from CRC metastases.