Advanced peritoneal carcinomatosis including high-grade ovarian cancer has poor prognoses and a poor response rate to current checkpoint inhibitor immunotherapies; thus, there is an unmet need for effective therapeutics that would provide benefit to these patients. Here we present the preclinical development of SENTI-101, a cell preparation of bone marrow-derived mesenchymal stromal (also known as stem) cells (MSC), which are engineered to express two potent immune-modulatory cytokines, IL12 and IL21. Intraperitoneal administration of SENTI-101 results in selective tumor-homing and localized and sustained cytokine production in murine models of peritoneal cancer. SENTI-101 has extended half-life, reduced systemic distribution, and improved antitumor activity when compared with recombinant cytokines, suggesting that it is more effective and has lower risk of systemic immunotoxicities. Treatment of tumor-bearing immune-competent mice with a murine surrogate of SENTI-101 (mSENTI-101) results in a potent and localized immune response consistent with increased number and activation of antigen presenting cells, T cells and B cells, which leads to antitumor response and memory-induced long-term immunity. Consistent with this mechanism of action, co-administration of mSENTI-101 with checkpoint inhibitors leads to synergistic improvement in antitumor response. Collectively, these data warrant potential clinical development of SENTI-101 for patients with peritoneal carcinomatosis and high-grade ovarian cancer.

Graphical abstract: SENTI-101 schematic and mechanism of action

SENTI-101 is a novel cell-based immunotherapeutic consisting of bone marrow–derived mesenchymal stromal cells (BM-MSC) engineered to express IL12 and IL21 intended for the treatment of peritoneal carcinomatosis including high-grade serous ovarian cancer. Upon intraperitoneal administration, SENTI-101 homes to peritoneal solid tumors and secretes IL12 and IL21 in a localized and sustained fashion. The expression of these two potent cytokines drives tumor infiltration and engagement of multiple components of the immune system: antigen-presenting cells, T cells, and B cells, resulting in durable antitumor immunity in preclinical models of cancer.

This article is featured in Highlights of This Issue, p. 1497

Multiple solid tumors, including gynecological and gastrointestinal tumors, have the capacity to metastasize and spread through the peritoneal cavity in a process known as peritoneal carcinomatosis (PC; ref. 1). One of the most frequent tumors that metastasize to the peritoneal cavity is advanced high-grade serous ovarian cancer (HGSOC; refs. 2, 3), which accounts for more than 20,000 new cases every year in the United States alone and is often associated with disease progression and low overall survival rates (<30% 5-year survival rate; ref. 4).

Current treatment approaches for HGSOC include cytoreductive debulking surgery combined with platinum-based chemotherapy, including intraperitoneal chemotherapy (5, 6). In the last year, PARP inhibitors have been approved as a second-line maintenance treatment for HGSOC (7, 8). Although current standard of care therapies prolong time to progression, they have limited impact on overall survival (9, 10). Thus, novel therapeutics are needed to address this unmet medical need.

The remarkable success of immunotherapy in the treatment of certain solid tumors, such as melanoma or lung cancer (11), has not effectively translated into the treatment of advanced HGSOC; recent clinical trials have shown a disappointing lower response rate (12–15) and, hence, an efficacious and durable response to treatment remains elusive for these patients. There have been clinical studies in HGSOC that attempted to alternatively engage the patient's immune system by using immune-stimulatory cytokines such as IL12 (16–20). Although cytokines are powerful modulators of the immune system, their therapeutic potential is limited by their short half-lives and systemic toxicities (21, 22).

A novel therapeutic modality that is currently being explored for the treatment of HGSOC consists of mesenchymal stromal cells [also referred to as mesenchymal stem cells (MSCs); refs. 23–26]. MSCs are a heterogeneous and undifferentiated cell population that can be sourced from bone marrow (BM-MSC), umbilical cord (UC-MSC), and adipose tissues (Ad-MSC), and have the ability to differentiate into osteoblast, adipocyte, and chondroblast lineages upon appropriate stimulation (25–26). MSCs are immune-privileged and lack the expression of HLA-II molecules, making them an attractive candidate for off-the-shelf allogeneic cell therapies (27–30). Furthermore, BM-MSCs have innate tumor-homing capacity (31, 32) and are known to preferentially interact with tumor extracellular matrix components (33), thus having the potential to selectively deliver potent drugs to the tumor microenvironment (34–36).

Here we present the preclinical development of SENTI-101, an allogeneic cell-based therapeutic consisting of human BM-MSCs engineered to overexpress two potent immune-stimulatory cytokines, IL12 and IL21, resulting in a multimodal and durable antitumor immune response in peritoneal solid tumors.

MSC culture

Murine MSCs were purchased from Cyagen (Balb/c MSCs #MUCMX-01001; C57Bl/6 MSCs #MUBMX-01001). Human bone marrow-derived MSCs were obtained from Rooster Bio (RUO and GMP-grade donors). MSCs were passaged at 70% to 90% confluency. Cells were trypsinized using TrypLE and subcultured at 3,000 to 5,000 cells/cm2. Murine MSCs were cultured using minimum essential medium (MEM) alpha medium containing 10% MSC-FBS, 200 mmol/L L-glutamine, 1 μg/μL of murine FGF-basic, and penicillin/streptomycin. Cells were confirmed free of mycoplasma and tested routinely. Human MSCs were cultured with RoosterBasal-MSC and RoosterBooster-MSC-XF Supplement. Cells were passaged for a limited number of times (<6 passages) and were resuspended in 200 μL of PBS for in vivo administration at the doses listed in each figure legend.

Lentiviral vector production and MSC transduction

Lenti-X 293T packaging cell line was cultured in DMEM, high-glucose, 1 mmol/L sodium pyruvate, 10% heat-inactivated fetal bovine serum (HI-FBS) and seeded at 72,700 cells/cm2 prior to transfection. Lentiviral transfer vectors were cotransfected with psPAX2 packaging vector and pMD2.g envelope vector containing the VSV-G envelope protein using FuGene-HD. Viral supernatants were concentrated using Lenti-X concentrator. Concentrated vectors were added to MSCs seeded in 6-well plates 4 hours prior to viral transduction. Cells were spinoculated at 800 g for 1 hour at 32°C, followed by incubation at 37°C 5% CO2. Four hours post-spinoculation, an additional 2 mL of complete media was added to each well. Control MSCs were generated in parallel to vector-engineered MSCs by transducing the cells with a sham lentiviral vector without the expression cassette.

In vivo murine models of peritoneal carcinomatosis

CT26.WT-Fluc-Neo were purchased from Imantis Life Sciences (Catalog no.: CL043; Lot no.: CL-IM147) and were cultured in DMEM media with 10% FBS and 0.4 mg/mL G418. B16F10-Fluc-Puro were purchased from Imantis Life Sciences (Catalog no.: CL052; Lot no.: CL-IM150) and cultured in RPMI media with 10% FBS and 1 μg/mL puromycin. Either 5e4 (CT26) or 1e5 (B16F10) cells were injected intraperitoneally into female Balb/c or C57Bl/6J mice (The Jackson Laboratory). Tumor burden was measured by bioluminescence imaging after injection of D-Luciferin substrate (15 mg/mL). For in vivo and ex vivo imaging of reporter cells (transduced with the nanoLuc reporter) 100 μL of nanoGlo substrate solution was injected into mice, and mice or tissues were immediately imaged. AMI and Lago imagers (Spectral Instruments) were used with prior calibration and appropriate negative controls. AMI software was used to quantify bioluminescence flux (photons/second) in a region of interest (ROI). Subcutaneous tumor volume was measured using a caliper.

Tumor burden, body weight, and mice overall health were assessed weekly, and mice were euthanized at scheduled time points or when reaching endpoint criteria for survival studies. On scheduled time points tissues were collected as follows: blood was collected in EDTA-coated tubes via submandibular bleeding or intracardiac puncture and processed for flow cytometry or collected in serum (clot activator) tubes and centrifuged for serum separation; peritoneal fluid was collected via peritoneal lavage; peritoneal tumors and other organs were collected, and flash frozen in liquid N2 or fixed in formalin and embedded in paraffin for histologic evaluation.

All mouse studies were performed in compliance with Institutional Animal Care and Use Committee guidelines (IACUC protocol EB17-010-108).

IL12 reporter

HEK-Blue IL12 cells, an IL12 reporter cell line capable of sensing both mouse and human variants of IL12 (hkb-il12, from InvivoGen), were purchased and used used following manufacturer's instructions. Supernatants collected from MSC cultures were centrifuged at 400 × g for 5 minutes to remove cells prior to assay. HEK-Blue IL12 cells were seeded with sample supernatants in a 96-well plate, which were then incubated for 24 hours. A total of 180 μL of QUANTI-Blue were mixed with 20 mL of supernatants collected from the HEK-Blue IL12 cells. Absorbance was measured at 650 nm using BioTek SynergyH1 spectrophotometer. Recombinant mIL-12, mIL-21 and culture media were used as positive and negative controls for this assay.

Phospho-flow (pSTAT1 and pSTAT3)

Supernatants from MSC cultures or control samples were added in a 24-well plate with NK-92 cells. Cells were incubated for 30 minutes and fixed and permeabilized using Cytofix/Cytoperm Kit. Cells were stained with fluorescently labeled antibodies for pSTAT1 or pSTAT3. Recombinant IL12 (10 ng/mL) or IL21 (10 ng/mL) were used as negative or positive controls, respectively. Samples were analyzed using Cytoflex LX flow cytometer (Beckman Coulter) and Ultra-Bright counting beads for normalization.

PathHunter cytokine receptor assay

Assay was performed by Eurofins DiscoverX using a reporter cell line (PathHunter U2OS IL-21R/IL-2RG Dimerization Cell Line) in which one cytokine receptor chain is tagged with a small peptide epitope ProLink (PK) and the other chain is tagged with enzyme acceptor (EA). Ligand (IL21) binding induces dimerization of the two receptors, facilitating complementation of PK and EA fragments. This interaction generates an active unit of β-galactosidase, which is detected using a chemiluminescent substrate.

Protein concentration determination using ELISA and multiplex immunoassay

MSCs were seeded in a 6-well plate with 2 mL of media and supernatants collected at 24 hours. Mouse and human ELISA kits for IL12 and IL21 were purchased from R&D Systems and were used following manufacturer's instructions. Plates were read at 540 nm (reference wavelength) and 450 nm using BioTek SynergyH1 microplate reader spectrophotometer and Gen5 software. 10-plex Murine ProcartaPlex Luminex kits were customized by Thermo Fisher Scientific(Assay ID MXXGRM9; Life Technologies) and used following manufacturer's instructions. Primary murine tissue samples (serum or intraperitoneal fluid) were diluted in sample assay buffer where multiple dilutions were used to ensure all analytes were within standard range. Samples that required less dilution (<10-fold) were diluted in ProcartaPlex Platinum Assay Buffer (Thermo Fisher Scientific, Catalog No. EPXP-1112-000) to mitigate bead aggregation, whereas more concentrated samples that required further dilution (>10-fold) were diluted with the universal assay buffer provided in the kit. Human IL12 and IL21 levels in xenograft models were quantified with MilliPlex (EMD Millipore) kits following manufacturer's instructions. Samples were analyzed in Prism 8 using a weighted-5PL curve fit of the standards to interpolate sample concentrations.

Antibody depletion and treatment with anti-PD1 antibodies

Mice were depleted of CD8, CD4, and NK1.1+ cells using 200 μg in 200 μL of depleting antibodies administered intraperitoneally every 3 days for a total of four doses. One week after start of the treatment, immune cell depletion was confirmed by collecting blood via submandibular bleeding and performing flow cytometry to quantify CD4, CD8, and NK cells. Checkpoint inhibitor antibody anti-PD1, clone RMP1–14 was administered intraperitoneally in three doses of 200 mg/kg every 3 days, starting 1 day prior to treatment with MSCs.

Tissue processing and flow cytometry

Mice were euthanized and tissues (tumor, spleen, peritoneal sacral and lumbar lymph nodes, peritoneal fluid, and blood) were collected. Tumors were digested using MACS Mouse Tumor Dissociation Kit and the gentle MACS Octo Dissociator (Miltenyi Biotec). Single cell suspensions from all organs were obtained using a 70 μm strainer. Red blood cells were lysed using RBC lysis buffer. Cell suspensions were stained with fluorescently labeled antibodies (BioLegend and BD Biosciences, see supplementary table of reagents), adding Brilliant Stain Buffer. For intracellular staining, cells were permeabilized and fixed using the Foxp3 fixation and permeabilization kit following manufacturer's instructions. For tetramer staining, samples were minimally processed prior to incubation with H-2Ld MuLV gp70 Tetramer-SPSYVYHQF antibody (MBL) and membrane antibodies. Dead cells were excluded using FVS780-fixable viability dye (BD Biosciences). Samples were analyzed using the Cytoflex LX flow cytometer (Beckman Coulter) and FlowJo was used to analyze the data.

Multiplexed-IHC

Tumors from control and treated mice were fixed in formalin and embedded in paraffin for histology. Slides were deparaffinized in an oven chamber for 30 minutes at 37°C and then for 30 minutes at 60°C. Following deparaffinization, slides were rehydrated in Slide Brite, then in decreasing percentages of ethanol. After rehydration, antigen retrieval was performed using Rodent (pH 6) or Nuclear (pH 9.5) decloacker solution in NxGen Decloaking chamber at 95°C for 15 minutes. Slides were blocked using Rodent Block M and incubated for 30 minutes with primary antibodies, covered from light at RT. After primary antibody staining, slides were incubated with a secondary HRP-Polymer using the Opal 4-Color Anti-Rabbit Manual IHC Kit or Rat-on-Mouse HRP-Polymer for Rat antibodies (depending on the primary antibody), followed by opal fluorophore staining (Opal520, Opal570, or Opal690; Akoya Biosciences). DAPI staining was used to counterstain nuclei.

Universal Negative Control reagent was used instead of the primary antibody as a negative control. Slides were mounted using ProLong Diamond Antifade Mountant and scanned using Vectra Polaris microscope (Akoya Biosciences) and Phenochart software was used for visualization.

Detailed descriptions of reagents including catalog numbers are shown in Supplementary Materials Table.

Engineered MSCs home into peritoneal tumors and have antitumor activity in syngeneic mouse models with distinct immune landscapes

MSCs have been previously shown to home into sites of inflammation and tumors, including intraperitoneal tumors (31, 36). We confirmed that BM-MSCs engineered to express a bioluminescent reporter (firefly luciferase) preferentially localized into tumor tissues compared with other peritoneal healthy organs when delivered intraperitoneally (Fig. 1A and B; Supplementary Figs. S1A and S1B). In terms of kinetics, IP-delivered MSCs were detected up to 3 days after administration, whereas the signal decreased by day 10 in syngeneic models of peritoneal carcinomatosis (B16-F10 model; Supplementary Fig. S1C). The innate tumor-tropism and kinetics of MSCs makes them an ideal candidate to deliver potent immunomodulatory agents to the tumor microenvironment, aimed to locally polarize and activate the immune system and thus avoid potential systemic immunotoxicities. To further evaluate this novel MSC-based immunomodulatory therapy concept, we engineered murine (Balb/c and C57Bl/6) BM-MSCs to express different payloads that have known immune stimulatory function (including cytokines, chemokines, and tumor microenvironment modifiers) and empirically determined their abilities to induce antitumor responses as single agents or in combination using two different syngeneic mouse models of intraperitoneal carcinomatosis with distinct immune landscapes: CT26-ip and B16F10-ip (37, 38). As part of the characterization of these syngeneic IP tumor models, we analyzed their immune infiltrates by flow cytometry and IHC (Fig. 1C; Supplementary Figs. S2A and S2B). Control-MSCs (engineered with a sham or nonexpression lentiviral vector) were used as control. Antitumor activity of different combinations was assessed by comparing the tumor burden fold change 10 days after treatment. Extensive in vivo screening of over 20 payloads (cytokines, chemokines, and immune activators) with different mechanisms of action tested either alone or in combinations in both syngeneic immunocompetent IP tumor models (CT26-ip and B16F10-ip). Although multiple combinations achieved significant tumor burden reduction in the CT26-ip model that contains more T-cell infiltrates (Fig. 1D; refs. 36, 37), the immune refractory B16F10-ip model, revealed that the combination of IL12 and IL21 resulted in the most robust antitumor response (Fig. 1E; Supplementary Fig. S3). IL12 and IL21 are potent immune-stimulatory cytokines with multimodal immune modulatory mechanisms of action and have previously been shown to demonstrate antitumor activities in multiple nonclinical models of cancer (IL12; refs. 39–42), IL21; refs. 43–46) and in the clinic (17–21). This combination was selected as the candidate, SENTI-101.

Figure 1.

Intraperitoneally delivered MSCs home into solid tumors and are a platform to locally deliver immune-modulatory factors. A, Mice were injected with murine cancer cells (CT26) into the peritoneal space. Murine Balb/c BM-MSCs engineered to express the bioluminescent reporter firefly Luciferase (fLUC) and were injected intraperitoneally on day 7 posttumor implant. Mice were euthanized 24 hours after MSC injection and tumors and organs in the peritoneal space (pancreas, liver, kidney, spleen, and omentum) were collected and imaged ex vivo after addition of the substrate luciferin. fLUC bioluminescence was quantified using IVIS. B, MSCs in CT26 tumors were imaged using immunofluorescence microscopy to detect fLuciferase protein. DAPI was used to counterstain nuclei. C, Two preclinical models of peritoneal carcinomatosis were established in immunocompetent mice using CT26-fLUC colorectal carcinoma cells (Balb/c) and B16F10-fLUC melanoma cells (C57Bl/6). Tumor immune infiltrates (myeloid cells, T cells, and B cells) were profiled using multiplexed IHC and flow cytometry (Supplementary Fig. S2). D and E, Murine Balb/c or C57Bl/6 MSCs were engineered to express different immune modulators (cytokines, chemokines, immune stimulatory factors) and used to treat established CT26-ip (D) or B16F10-ip (E) peritoneal tumors either as single therapy or in combinations of two or three effectors. Tumor burden was measured using fLuciferase bioluminescence (BLI). Fold change in tumor burden was calculated comparing the BLI signal 10 days after treatment compared with the signal at randomization for each individual mouse. Each dot represents an individual mouse.

Figure 1.

Intraperitoneally delivered MSCs home into solid tumors and are a platform to locally deliver immune-modulatory factors. A, Mice were injected with murine cancer cells (CT26) into the peritoneal space. Murine Balb/c BM-MSCs engineered to express the bioluminescent reporter firefly Luciferase (fLUC) and were injected intraperitoneally on day 7 posttumor implant. Mice were euthanized 24 hours after MSC injection and tumors and organs in the peritoneal space (pancreas, liver, kidney, spleen, and omentum) were collected and imaged ex vivo after addition of the substrate luciferin. fLUC bioluminescence was quantified using IVIS. B, MSCs in CT26 tumors were imaged using immunofluorescence microscopy to detect fLuciferase protein. DAPI was used to counterstain nuclei. C, Two preclinical models of peritoneal carcinomatosis were established in immunocompetent mice using CT26-fLUC colorectal carcinoma cells (Balb/c) and B16F10-fLUC melanoma cells (C57Bl/6). Tumor immune infiltrates (myeloid cells, T cells, and B cells) were profiled using multiplexed IHC and flow cytometry (Supplementary Fig. S2). D and E, Murine Balb/c or C57Bl/6 MSCs were engineered to express different immune modulators (cytokines, chemokines, immune stimulatory factors) and used to treat established CT26-ip (D) or B16F10-ip (E) peritoneal tumors either as single therapy or in combinations of two or three effectors. Tumor burden was measured using fLuciferase bioluminescence (BLI). Fold change in tumor burden was calculated comparing the BLI signal 10 days after treatment compared with the signal at randomization for each individual mouse. Each dot represents an individual mouse.

Close modal

SENTI-101 is a preparation of hBM-MSCs engineered to express IL12 and IL21

To optimize the expression of IL12 and IL21 in human BM-MSCs, we evaluated a library of commonly used promoters to drive the expression of an EGFP reporter in our lentiviral transfer vector backbone. hBM-MSCs were transduced with a lentiviral vectors encoding different promoters driving a fluorescent reporter protein, EGFP. To select the best promoters from this panel, transduction efficiency was evaluated using reporter fluorescence intensity as measured from flow cytometry (Fig. 2A and B; Supplementary Fig. S4A). In parallel, constructs with IL12 and IL21 were generated using spleen focus-forming virus (SFFV) or elongation factor-1 alpha (EF1a) promoters and cytokine production was compared (Fig. 2C). In both instances, SFFV promoter outperformed other promoters such as EF1a or EFS in driving robust and durable expression of the expression cassette for multiple passages and was selected for our construct. Furthermore, we tested different orientations as well as the presence of different signal sequence peptides to enhance the secretion of these cytokines (Fig. 2D; Supplementary Fig. S4B). Protein function and signaling was confirmed for each cytokine using flow-based and cell-based reporter assays to detect induction of STAT phosphorylation and subsequent activation of downstream signals. All assays confirmed that our engineered MSCs secrete functional IL12 and IL21 cytokines (Fig. 2E and F; Supplementary Fig. S5). Although there was a modest increase in IL21 protein secretion by using IL8 signal sequence (construct SB00971), the difference was not sufficient to warrant such modification, and thus we selected the construct with the naïve codon-optimized sequences of IL12p70 single chain and IL21 linked with a T2A self-cleaving peptide (construct SB00880) as our lead candidate for manufacturing and development of SENTI-101 [Fig. 2G; Supplementary Fig. S6; Supplementary Table S1, United States Patent Application 2020017109; United States Patent Publication No. 2020/0206271; United States Patent No. 10,993,967 (pending) 47].

Figure 2.

Disclosure of SENTI-101. Lead candidate optimization, functional validation and construct map. A and B, Human BM-MSCs from two independent donors were engineered with constructs in which different promoters were driving EGFP expression. EGFP fluorescence was quantified using flow cytometry. % transduction (A) and mean fluorescence intensity (MFI; B) were compared across multiple cell lines and through multiple passages (up to day 28 post-transduction). C, Human BM-MSCs were engineered to overexpress the lead candidate of cytokines IL12 and IL21. SFFV or EF1α were the promoters driving protein expression. Cytokine production (pg/1e6 cells) was quantified by ELISA in supernatants collected 24 hours after seeding the cells. Results are shown as average of technical replicates (N = 3). D, Different constructs were designed to enhance cytokine production by changing the signal sequence of IL21 (naïve compared with no signal sequence, IL2 signal sequence or IL8 signal sequence). Cytokine production of IL12 or IL21 (pg/1e6 cells) was quantified by ELISA in supernatants collected 24 hours after seeding the cells. Results are shown as average and SEM of independent replicates (N = 3). E, IL12 function was confirmed using a reporter cell line (IL12-HEK-Blue). Reporter cells were incubated with supernatants collected from different MSC engineered with the listed constructs. Recombinant cytokines were used as control. Results are shown as average of 4 technical replicates. F, IL21 function was confirmed using phospho-flow for the downstream targets pSTAT1 and pSTAT3. NK92 cells were incubated with supernatant from different MSCs engineered with the listed constructs and relative expression of pSTAT1 or pSTAT3 was quantified. Recombinant cytokines were used as control. Results are shown as average of technical replicates. G, Plasmid map of SB00880, our selected lead candidate construct for SENTI-101. The construct encodes for IL12 and IL21 human sequences codon-optimized linked by a furin-T2A sequence and under the SFFV promoter and contains a Woodchuck hepatitis virus (WHP) posttranscriptional regulatory element (WPRE) to enhance expression.

Figure 2.

Disclosure of SENTI-101. Lead candidate optimization, functional validation and construct map. A and B, Human BM-MSCs from two independent donors were engineered with constructs in which different promoters were driving EGFP expression. EGFP fluorescence was quantified using flow cytometry. % transduction (A) and mean fluorescence intensity (MFI; B) were compared across multiple cell lines and through multiple passages (up to day 28 post-transduction). C, Human BM-MSCs were engineered to overexpress the lead candidate of cytokines IL12 and IL21. SFFV or EF1α were the promoters driving protein expression. Cytokine production (pg/1e6 cells) was quantified by ELISA in supernatants collected 24 hours after seeding the cells. Results are shown as average of technical replicates (N = 3). D, Different constructs were designed to enhance cytokine production by changing the signal sequence of IL21 (naïve compared with no signal sequence, IL2 signal sequence or IL8 signal sequence). Cytokine production of IL12 or IL21 (pg/1e6 cells) was quantified by ELISA in supernatants collected 24 hours after seeding the cells. Results are shown as average and SEM of independent replicates (N = 3). E, IL12 function was confirmed using a reporter cell line (IL12-HEK-Blue). Reporter cells were incubated with supernatants collected from different MSC engineered with the listed constructs. Recombinant cytokines were used as control. Results are shown as average of 4 technical replicates. F, IL21 function was confirmed using phospho-flow for the downstream targets pSTAT1 and pSTAT3. NK92 cells were incubated with supernatant from different MSCs engineered with the listed constructs and relative expression of pSTAT1 or pSTAT3 was quantified. Recombinant cytokines were used as control. Results are shown as average of technical replicates. G, Plasmid map of SB00880, our selected lead candidate construct for SENTI-101. The construct encodes for IL12 and IL21 human sequences codon-optimized linked by a furin-T2A sequence and under the SFFV promoter and contains a Woodchuck hepatitis virus (WHP) posttranscriptional regulatory element (WPRE) to enhance expression.

Close modal

Pharmacokinetics and pharmacodynamics of SENTI-101

To characterize the pharmacokinetic (PK) and pharmacodynamic (PD) parameters of SENTI-101, we used a murine surrogate, mSENTI-101 (murine BM-MSCs engineered to express the equivalent dose of murine IL12 and IL21; Supplementary Fig. S7A). We delivered mSENTI-101 via intraperitoneal injection into syngeneic immunocompetent tumor-bearing mice (CT26-ip and B16F10-ip). A bolus dose of recombinant cytokines was injected in a separate group of mice. The dose of recombinant cytokines was four times the daily dose produced by mSENTI-101 in vitro to account for the sustained production over approximately 4 days by the engineered MSCs in vivo. PF and serum were collected at multiple time points, and cytokine concentrations of IL12 and IL21 as well as cytokines and chemokines that were predicted to be downstream of IL12 and IL21 were determined via Luminex. IL12 cytokine concentration in locoregional and systemic compartments was measured and the PKs of recombinant cytokines and Senti-101 were nonlinearly fit with a weighted (1/y) single-phase exponential decay using GraphPad Prism. The same nonlinear regression was applied to both treatment arms. (Fig. 3A; Supplementary Fig. S7B). SENTI-101 administration resulted in a more durable and localized presence of IL12 compared with administration of recombinant cytokines. Half-life of IL12 in the mSENTI-101 group was more than four times longer compared with the recombinant cytokines (10.39 hours for mice treated with mSENTI-101 vs. 2.3 hours for mice treated with recombinant cytokines; Supplementary Fig. S7B). Importantly, the ratio between PF and serum levels of IL12 was 10-fold lower in mice treated with SENTI-101 compared with the treatment with recombinant cytokines (Fig. 3B). The limited systemic exposure of IL12 and IL21 observed with SENTI-101 treatment offers an important advantage over recombinant cytokines, reducing the potential risk for systemic immunotoxicities that has been previously observed with IL12 (18, 23).

Figure 3.

PK and PD of SENTI-101. A, Balb/c murine SENTI-101 (mSENTI-101) MSCs were injected intraperitoneally into mice harboring established CT26-ip tumors. In parallel, a separate cohort of mice was treated with recombinant murine IL12 and IL21 in a bolus dose four times higher than the amount of IL12 and IL21 being produced by mSENTI-101 measured by ELISA. Peritoneal fluid and serum were collected at the listed time points after MSC injection and cytokine concentration (murine IL12) was measured by Luminex. Darker color indicates the cytokine measurements from mice treated with mSENTI-101 and lighter color indicates mice treated with recombinant murine cytokines. Each dot represents an individual mouse. Control MSCs (engineered with a non-coding construct) and mice treated with PBS were used as control (baseline indicated by gray-shaded area). B, Ratio between serum and peritoneal fluid concentration of IL12, comparing the treatment with mSENTI-101 to the treatment with murine cytokines. C, Kaplan–Meier survival curve of mice harboring CT26-ip tumors treated with mSENTI-101 (dark teal) or with recombinant murine cytokines as described in a. PBS and control MSCs were used as control. D, CT26-ip or B16F10-ip tumor-bearing mice were treated with mSENTI-101 or control MSCs and peritoneal fluid was collected at different time points after treatment. Ten cytokines and chemokines (IL21, IFNγ, IP-10/CXCL-10, TNFα, IL2, IL2Ra/sCD25, IL6, IL10, RANTES/CXCL-9 in addition to IL12p70) were measured using Luminex. Heat-maps showing the relative expression of cytokines and chemokines that are induced after treatment with mSENTI-101 in the CT26 (left) or in the B16F10 (right) tumor models as a function on time (on the x-axis). Data normalized to maximum and minimum for each analyte. Fold-change over baseline (control MSCs) values are shown in Supplementary Fig. S7F. CT26-ip model: N = 4 mice per treatment arm per time point (3, 24, 48, and 72 hours) and N = 7 mice per group for survival. B16F10-ip model: N = 4 mice per treatment arm and per time point (3, 24, 48, and 72 hours, 7 days and survival).

Figure 3.

PK and PD of SENTI-101. A, Balb/c murine SENTI-101 (mSENTI-101) MSCs were injected intraperitoneally into mice harboring established CT26-ip tumors. In parallel, a separate cohort of mice was treated with recombinant murine IL12 and IL21 in a bolus dose four times higher than the amount of IL12 and IL21 being produced by mSENTI-101 measured by ELISA. Peritoneal fluid and serum were collected at the listed time points after MSC injection and cytokine concentration (murine IL12) was measured by Luminex. Darker color indicates the cytokine measurements from mice treated with mSENTI-101 and lighter color indicates mice treated with recombinant murine cytokines. Each dot represents an individual mouse. Control MSCs (engineered with a non-coding construct) and mice treated with PBS were used as control (baseline indicated by gray-shaded area). B, Ratio between serum and peritoneal fluid concentration of IL12, comparing the treatment with mSENTI-101 to the treatment with murine cytokines. C, Kaplan–Meier survival curve of mice harboring CT26-ip tumors treated with mSENTI-101 (dark teal) or with recombinant murine cytokines as described in a. PBS and control MSCs were used as control. D, CT26-ip or B16F10-ip tumor-bearing mice were treated with mSENTI-101 or control MSCs and peritoneal fluid was collected at different time points after treatment. Ten cytokines and chemokines (IL21, IFNγ, IP-10/CXCL-10, TNFα, IL2, IL2Ra/sCD25, IL6, IL10, RANTES/CXCL-9 in addition to IL12p70) were measured using Luminex. Heat-maps showing the relative expression of cytokines and chemokines that are induced after treatment with mSENTI-101 in the CT26 (left) or in the B16F10 (right) tumor models as a function on time (on the x-axis). Data normalized to maximum and minimum for each analyte. Fold-change over baseline (control MSCs) values are shown in Supplementary Fig. S7F. CT26-ip model: N = 4 mice per treatment arm per time point (3, 24, 48, and 72 hours) and N = 7 mice per group for survival. B16F10-ip model: N = 4 mice per treatment arm and per time point (3, 24, 48, and 72 hours, 7 days and survival).

Close modal

Consistent with the results obtained in the CT26-ip model, mSENTI-101 resulted in a peritoneal localized and sustained production of IL12 and IL21 with a longer half-life when compared with recombinant cytokines in B16F10-ip models (Supplementary Figs. S7C and S7D). Of note, there were differences in the kinetics and cytokine systemic exposure between CT26-ip and B16F10-ip models, which can be due to different baseline immune landscapes present in the tumors, as well as different mouse backgrounds (Balb/c vs. C57Bl/6; refs. 37, 38, 48). Consistent with increased cytokine half-life, mSENTI-101 treatment resulted in increased antitumor activity and proportion of tumor-free survival in tumor-bearing mice compared with recombinant cytokines (Fig. 3C; Supplementary Fig. S7E).

IL12 and IL21 have pleiotropic and non-overlapping roles, but combination of both results in activation of T and NK cells, which lead to IFNγ production (39, 49). We quantified the levels of IFNγ as well as other IFN-induced chemokines such as IP10, as part of our PD panel. We observed a significant and durable increase in multiple immune-stimulatory factors (IFNγ, IP10, sCD25) as a result of the treatment with mSENTI-101, indicative of a robust antitumor immune response (Fig. 3D). Our panel also included other inflammatory cytokines typically involved in immune-mediated toxicities and cytokine release syndrome (CRS) such as TNFα or IL6. Although in mice treated with mSENTI-101, we observed relative increases in such cytokines in the early time points (3 hours), these increases were minor when compared with baseline (Supplementary Fig. S7D).

SENTI-101 induces antitumor immunity

Recombinant IL12 and IL21 have each been evaluated for the treatment of cancer due to its immune stimulatory functions, which can result in enhanced antitumor immunity (39, 43, 44). Importantly, several strategies to safely and locally deliver IL12 have been tested in clinical studies for cancer (NCT04006119, NCT03393884). To test for antitumor activity of our lead candidate, we used the murine surrogate (mSENTI-101) in both CT26-ip and B16F10-ip models. Immune competent mice with established syngeneic tumors were treated on day 7 posttumor implant with IP-delivered engineered MSCs (Supplementary Fig. S8A). Treatment with mSENTI-101 resulted in a significant and dose-dependent reduction in tumor burden as measured by bioluminescence imaging (Fig. 4A and B). This included the complete eradication of over 90% of the tumors in both models 10 days after treatment (Fig. 4C and D; Supplementary Figs. S8B and S8C). Importantly, in the CT26-ip tumor model that develops ascites, treatment with mSENTI-101 significantly reduced the presence of malignant ascites (Fig. 4E), which is an important feature of peritoneal carcinomatosis and advanced ovarian cancer (2, 3). None of the doses of SENTI-101 resulted in any toxicities or decreased body weight in the mice (Supplementary Fig. S9).

Figure 4.

Antitumor activity of SENTI-101 in syngeneic preclinical models of peritoneal carcinomatosis. A, Balb/c mice with established CT26-ip tumors were treated with decreasing doses (shown in the figure) of Balb/c mSENTI-101. Tumor burden was measured via fLuciferase bioluminescence. Fold change in tumor burden was calculated comparing the tumor burden quantification 10 days after treatment to the tumor burden prior to treatment for each individual mouse. Mice were treated with PBS or with control MSCs (engineered with a lentiviral construct without an expression cassette) as control. Each dot represents an individual mouse. Representative images of the fLuciferase bioluminescence on day 10 after treatment are shown. B, C57Bl/6 mice with established B16F10-ip tumors were treated with decreasing doses (shown in the figure) of C57Bl/6 mSENTI-101. Tumor burden was measured via fLuciferase bioluminescence. Fold change in tumor burden was calculated comparing the tumor burden quantification 10 days after treatment to the tumor burden prior to treatment for each individual mouse. Mice were treated with PBS or with control MSCs (engineered with a lentiviral construct without an expression cassette) as control. Each dot represents an individual mouse. N = 8 mice per treatment group (CT26-ip) or N = 10 mice per treatment group (B16F10-ip). Representative images of the fLuciferase bioluminescence on day 10 after treatment are shown. C and D, Tumor weights at termination (10 days after treatment) on the CT26-ip (C) or B16F10-ip (D) tumor model. Tumor bearing mice were treated with the effective dose of mSENTI-101 or the same dose of Control MSCs (1e6 MSCs were used in the CT26-ip model and 3e6 MSCs were used in the B16F10-ip model). CT26-ip: N = 18 mice per group from three independent experiments. B16F10-ip: N = 20 mice per group (PBS and control MSCs) or 13 mice (mSENTI-101), four independent experiments. E, Presence of ascites was determined 72 hours after treatment in CT26-ip tumor-bearing animals after treatment with mSENTI-101 (1e6 cells/mouse). Control MSCs at the same dose or PBS were used as vehicle controls. N = 18 mice per group (PBS and control MSCs) or 20 mice (mSENTI-101). F, Balb/c mice were depleted of T cells (CD4, CD8) or natural killer (NK) cells using antibodies (experimental design and timeline shown in Supplementary Fig. S8). After immune cell depletion was confirmed by flow cytometry, CT26 tumors were established intraperitoneally and mice were treated with Balb/c MSCs (mSENTI-101 or control MSCs). Tumor burden was measured by fLuciferase BLI and fold change and tumor weights at termination (day 10 after treatment) are plotted. PBS treatment was used as control. Each dot represents an individual mouse (N = 5 mice per group). Representative images of fLuciferase imaging are shown. G, C57Bl/6 mice were depleted of CD4, CD8, or NK cells using antibodies. After immune cell depletion was confirmed by flow cytometry, B16F10 tumors were established intraperitoneally and mice were treated with C57Bl/6 MSCs (mSENTI-101 or control MSCs). Tumor burden was measured by fLuciferase BLI and fold change and tumor weights at termination (day 10 after treatment) are plotted. PBS treatment was used as control. Each dot represents an individual mouse (N = 5 mice per group). Representative images of fLuciferase imaging are shown.

Figure 4.

Antitumor activity of SENTI-101 in syngeneic preclinical models of peritoneal carcinomatosis. A, Balb/c mice with established CT26-ip tumors were treated with decreasing doses (shown in the figure) of Balb/c mSENTI-101. Tumor burden was measured via fLuciferase bioluminescence. Fold change in tumor burden was calculated comparing the tumor burden quantification 10 days after treatment to the tumor burden prior to treatment for each individual mouse. Mice were treated with PBS or with control MSCs (engineered with a lentiviral construct without an expression cassette) as control. Each dot represents an individual mouse. Representative images of the fLuciferase bioluminescence on day 10 after treatment are shown. B, C57Bl/6 mice with established B16F10-ip tumors were treated with decreasing doses (shown in the figure) of C57Bl/6 mSENTI-101. Tumor burden was measured via fLuciferase bioluminescence. Fold change in tumor burden was calculated comparing the tumor burden quantification 10 days after treatment to the tumor burden prior to treatment for each individual mouse. Mice were treated with PBS or with control MSCs (engineered with a lentiviral construct without an expression cassette) as control. Each dot represents an individual mouse. N = 8 mice per treatment group (CT26-ip) or N = 10 mice per treatment group (B16F10-ip). Representative images of the fLuciferase bioluminescence on day 10 after treatment are shown. C and D, Tumor weights at termination (10 days after treatment) on the CT26-ip (C) or B16F10-ip (D) tumor model. Tumor bearing mice were treated with the effective dose of mSENTI-101 or the same dose of Control MSCs (1e6 MSCs were used in the CT26-ip model and 3e6 MSCs were used in the B16F10-ip model). CT26-ip: N = 18 mice per group from three independent experiments. B16F10-ip: N = 20 mice per group (PBS and control MSCs) or 13 mice (mSENTI-101), four independent experiments. E, Presence of ascites was determined 72 hours after treatment in CT26-ip tumor-bearing animals after treatment with mSENTI-101 (1e6 cells/mouse). Control MSCs at the same dose or PBS were used as vehicle controls. N = 18 mice per group (PBS and control MSCs) or 20 mice (mSENTI-101). F, Balb/c mice were depleted of T cells (CD4, CD8) or natural killer (NK) cells using antibodies (experimental design and timeline shown in Supplementary Fig. S8). After immune cell depletion was confirmed by flow cytometry, CT26 tumors were established intraperitoneally and mice were treated with Balb/c MSCs (mSENTI-101 or control MSCs). Tumor burden was measured by fLuciferase BLI and fold change and tumor weights at termination (day 10 after treatment) are plotted. PBS treatment was used as control. Each dot represents an individual mouse (N = 5 mice per group). Representative images of fLuciferase imaging are shown. G, C57Bl/6 mice were depleted of CD4, CD8, or NK cells using antibodies. After immune cell depletion was confirmed by flow cytometry, B16F10 tumors were established intraperitoneally and mice were treated with C57Bl/6 MSCs (mSENTI-101 or control MSCs). Tumor burden was measured by fLuciferase BLI and fold change and tumor weights at termination (day 10 after treatment) are plotted. PBS treatment was used as control. Each dot represents an individual mouse (N = 5 mice per group). Representative images of fLuciferase imaging are shown.

Close modal

To confirm that this antitumor activity was mediated by the immune system, we used depletion antibodies to selectively decrease different immune components that are known to mediate antitumor immunity: namely CD4, CD8, and NK cells (50). Mice were treated with depleting antibodies weekly prior to tumor implantation and throughout the duration of treatment (Supplementary Fig. S10A). Flow cytometry was performed to assess the reduction of the targeted cells (Supplementary Figs. S10B and S10C). Once immune depletion was confirmed, mice were implanted with peritoneal tumors and treated with intraperitoneally-injected MSCs 7 days thereafter. mSENTI-101-driven antitumor activity was significantly reduced when different immune subsets were depleted (Fig. 4F and G), demonstrating that SENTI-101 antitumor activity does indeed rely on the endogenous immune system, and specifically requires T cells and NK cells at a minimum.

SENTI-101 treatment locally induces antitumor immune activity

To further characterize the immune response induced by SENTI-101, we profiled the immune composition of tumors, PF, peritoneal lymph nodes, and circulating PBMCs from treated mice. mSENTI-101 was used to treat mice with established CT26-ip or B16F10-ip tumors and tissues were collected at 72 hours posttreatment and immune system composition was analyzed via multiplexed-IHC and flow cytometry. Treatment with mSENTI-101 resulted in increased T- and B-cell infiltration in peritoneal tumors (Fig. 5A and B; Supplementary Figs. S11A–S11C). These T cells and B cells were localized in well-organized clusters, indicative of tertiary lymphoid structures that were particularly evident in the immune “cold” model B16F10-ip (Supplementary Figs. S11B and S11C). These tertiary lymphod structures have been associated with improved prognosis as well as response to checkpoint inhibitors in multiple tumor types, including ovarian cancer (51–55). This was accompanied by a significant increase in activated CD8+ T cell subsets in the PF (Fig. 5C; Supplementary Fig. S12A). T cells in the PF of mice treated with mSENTI-101 expressed two or more cytokines (Fig. 5D; Supplementary Fig. S12B), suggestive of a stronger T-cell function, which has been shown to correlate with improved antitumor functions (56, 57). Consistent with an increase in IFNγ secretion, T cells in the peritoneal fluid expressed exhaustion markers such as Lag3 and PD1 (Supplementary Fig. S12C). Concomitantly, we observed a significant increase in antigen-presenting dendritic cells in the peritoneal tumor-draining lymph nodes (Fig. 5E; Supplementary Fig. S12D). Antigen-presenting dendritic cells (DC1 and DC2) are critical for durable and sustained antitumor activity and play a role in initiating the cancer immunity cycle (58–61). We also observed a reduction in myeloid derived suppressive cells (MDSC) in the blood of mice treated with mSENTI-101 (Supplementary Fig. S12E). Consistent with the localized cytokine production, the extent of the immune activation was much greater in the PF and tumors compared compared to circulation, with T-cell increases of 4- to 5-fold in tumor and PF of mice treated with mSENTI-101 compared with control-treated mice, whereas the increase in T cells was much lower in the periphery (<1.5-fold in treated mice compared with control; Fig. 5F; Supplementary Fig. S12F). These data support that treatment with SENTI-101 induces a potent but localized antitumor immune response, thus reducing the risk for systemic immune-mediated toxicities.

Figure 5.

Antitumor immune response induced by SENTI-101. A and B, Multiplex IHC was used to detect T cell (CD3) and B cell (CD19) immune infiltration into tumors. Balb/c mice with established CT26-ip tumors (A) or C57Bl/6 mice with established B16F10-ip tumors (B) were treated with mSENTI-101 via intraperitoneal injection. The same dose of control MSCs were used in a separate cohort as controls. Tumors were collected 72 hours after treatment and fixed in formalin and paraffin embedded to section for IHC staining. C, Peritoneal fluid from the same mice (B) treated with control or mSENTI-101 MSCs was collected via peritoneal lavage. Multiple panels of multicolor flow cytometry were used to quantify different immune subsets including activation and other functional markers. Each dot represents an individual mouse (N = 10 mice per group). D, Intracellular flow cytometry was used to quantify cytokines (IL2, TNFα, and IFNγ) on T cells, which were classified on the basis of the number of simultaneously expressed cytokines (1, 2, or 3). E, Peritoneal sacral and lumbar lymph nodes form the same mice (B) were collected and processed for flow cytometry. Antigen-presenting cell subsets and activation markers were quantified. Each dot represents an individual mouse. F, CD3 T cells quantified by flow cytometry in tumors, peritoneal fluid (PF) and blood of B16F10-ip tumor-bearing mice after treatment with mSENTI-101 or control MSCs. Median fold change calculated comparing the group treated with mSENTI-101 to the control group. Similar results using the CT26-ip tumor model are shown in Supplementary Fig. S12.

Figure 5.

Antitumor immune response induced by SENTI-101. A and B, Multiplex IHC was used to detect T cell (CD3) and B cell (CD19) immune infiltration into tumors. Balb/c mice with established CT26-ip tumors (A) or C57Bl/6 mice with established B16F10-ip tumors (B) were treated with mSENTI-101 via intraperitoneal injection. The same dose of control MSCs were used in a separate cohort as controls. Tumors were collected 72 hours after treatment and fixed in formalin and paraffin embedded to section for IHC staining. C, Peritoneal fluid from the same mice (B) treated with control or mSENTI-101 MSCs was collected via peritoneal lavage. Multiple panels of multicolor flow cytometry were used to quantify different immune subsets including activation and other functional markers. Each dot represents an individual mouse (N = 10 mice per group). D, Intracellular flow cytometry was used to quantify cytokines (IL2, TNFα, and IFNγ) on T cells, which were classified on the basis of the number of simultaneously expressed cytokines (1, 2, or 3). E, Peritoneal sacral and lumbar lymph nodes form the same mice (B) were collected and processed for flow cytometry. Antigen-presenting cell subsets and activation markers were quantified. Each dot represents an individual mouse. F, CD3 T cells quantified by flow cytometry in tumors, peritoneal fluid (PF) and blood of B16F10-ip tumor-bearing mice after treatment with mSENTI-101 or control MSCs. Median fold change calculated comparing the group treated with mSENTI-101 to the control group. Similar results using the CT26-ip tumor model are shown in Supplementary Fig. S12.

Close modal

SENTI-101 treatment results in the achievement of long-term tumor-free survival and immune memory

An important hallmark of a successful antitumor immunotherapeutic is the achievement of durable responses and tumor-free survival. To this end, we conducted long-term survival studies using both CT26-ip and B16F10-ip tumor models treated with mSENTI-101. In both cases, treatment with mSENTI-101 resulted in long-term tumor-free survival (73% in CT26-ip and 60% in B16F10-ip tumor models; Fig. 6A and B; Supplementary Fig. S13). Of note, the B16F10 model is highly immune-suppressed and refractory to multiple immune-oncology drugs, including anti-PD1/PD-L1 antibodies (37, 38). Treatment with mSENTI-101 significantly outperformed anti-PD1 antibodies and increased the response rate to such treatment from 27% to 85% of mice being tumor-free after 100 days (Fig. 6B). As observed by flow cytometry, treatment with SENTI-101 resulted in the engagement of multiple compartments of the cancer immunity cycle, which should ultimately lead to the acquisition of antitumor immune memory. We quantified the presence of CD8 T-cell clones that recognize the tumor antigen H2a using a tetramer staining against the CT26 Tetramer. We observed a significant increase in the presence of tetramer positive T cells in the PF of mice 14 days after treatment with mSENTI-101 (Fig. 6C). Concomitantly, T-cell memory subsets were significantly increased (Fig. 6D). Antitumor immune memory can be confirmed by a re-challenge of treated and tumor-free mice with new implantation of tumor cells (Supplementary Fig. S13C). All mice that were confirmed tumor-free for more than 90 days after treatment with mSENTI-101 were rechallenged with new tumor cells implanted subcutaneously in the flank. With the CT26 model, all the survivor mice treated with mSENTI-101 were able to reject the newly implanted tumors, demonstrating a successful achievement of antitumor immune memory (Fig. 6E). With the B16F10 model, 11 of 13 of the surviving mice after treatment with mSENTI-101 (85%) were able to reject the newly implanted tumor cells and, in all the cases, there was a significant delay in tumor growth, suggestive of a partial or initial antitumor immune response (Fig. 6F) in the 2 remaining mice. Overall, these results confirm that SENTI-101 treatment results in a robust and durable antitumor immune response and immune memory in syngeneic models of peritoneal cancer.

Figure 6.

Treatment with mSENTI-101 out-performs and improves response to checkpoint inhibitor anti-PD1 resulting in long-term tumor-free survival and antitumor immune memory. A and B, Balb/c mice with established CT26-ip tumors (A) or C57Bl/6 mice with established B16F10-ip tumors (B) were treated with mSENTI-101 via intraperitoneal injection. The same dose of control MSCs were used in a separate cohort as controls. Three doses of 200 mg/kg of antibodies anti-murine PD-1 (RMP1–14) were given intraperitoneally in combination with control MSCs or mSENTI-101. Red triangle indicates the time of MSC treatment (day 7 posttumor implant) and the red line on the x-axis denotes the duration of anti-PD1 treatment (day 6, 9, 12 posttumor implant). Tumor burden and symptoms were monitored weekly and mice were euthanized when displaying symptoms of excessive tumor burden. Kaplan–Meier survival curves were constructed and median survival for each treatment group was calculated. Individual tumor growth curves are show in Supplementary Fig. S13. A, Control-MSC + αPD1 vs. mSENTI-101 (3E5): P = 0.0069; mSENTI-101 (1E4) vs. mSENTI-101 (1E4) + αPD1: P = 0.0012; Control-MSC + αPD1 vs. mSENTI-101 (1E4) + αPD1: P = 0.3 (n.s.). B, Control-MSC vs. mSENTI-101: P = 0.0006; Control-MSC + αPD1 vs. mSENTI-101: P = 0.0014; mSENTI-101 vs. mSENTI-101 + αPD1: P = 0.13 (n.s.). Number of mice per treatment group are shown in the survival curve legend. C, Mice harboring CT26-ip tumors were treated with 1e6 control MSCs or mSENTI-101 and peritoneal fluid and tumors were collected 10 days after treatment. Tumor-antigen specific T cells were stained using a CT26-specific tetramer antibody (MHC-H2Ld Gp70 SPSYVYHQF) and quantified using flow cytometry. Each dot represents an individual mouse. D, Flow cytometry was used to profile memory T cells on the same mice from C. E and F, Balb/c mice with established CT26-ip tumors (E) or C57Bl/6 mice with established B16F10-ip tumors (F) were treated with mSENTI-101 via intraperitoneal injection. Tumor burden was measured weekly by bioluminescence imaging of fLuciferase. Mice that were confirmed tumor-free more than 90 days after treatment with mSENTI-101 were re-challenged by subcutaneous implantation of parental tumor cells in the flank. A cohort of treatment-naïve mice were implanted with tumor cells in parallel to serve as control. Tumor volume was measured using a caliper. Mice were euthanized when tumor volume reached 1,500 mm3. Individual tumor growth curves are plotted for the CT26 (E) and B16F10 (F) tumors.

Figure 6.

Treatment with mSENTI-101 out-performs and improves response to checkpoint inhibitor anti-PD1 resulting in long-term tumor-free survival and antitumor immune memory. A and B, Balb/c mice with established CT26-ip tumors (A) or C57Bl/6 mice with established B16F10-ip tumors (B) were treated with mSENTI-101 via intraperitoneal injection. The same dose of control MSCs were used in a separate cohort as controls. Three doses of 200 mg/kg of antibodies anti-murine PD-1 (RMP1–14) were given intraperitoneally in combination with control MSCs or mSENTI-101. Red triangle indicates the time of MSC treatment (day 7 posttumor implant) and the red line on the x-axis denotes the duration of anti-PD1 treatment (day 6, 9, 12 posttumor implant). Tumor burden and symptoms were monitored weekly and mice were euthanized when displaying symptoms of excessive tumor burden. Kaplan–Meier survival curves were constructed and median survival for each treatment group was calculated. Individual tumor growth curves are show in Supplementary Fig. S13. A, Control-MSC + αPD1 vs. mSENTI-101 (3E5): P = 0.0069; mSENTI-101 (1E4) vs. mSENTI-101 (1E4) + αPD1: P = 0.0012; Control-MSC + αPD1 vs. mSENTI-101 (1E4) + αPD1: P = 0.3 (n.s.). B, Control-MSC vs. mSENTI-101: P = 0.0006; Control-MSC + αPD1 vs. mSENTI-101: P = 0.0014; mSENTI-101 vs. mSENTI-101 + αPD1: P = 0.13 (n.s.). Number of mice per treatment group are shown in the survival curve legend. C, Mice harboring CT26-ip tumors were treated with 1e6 control MSCs or mSENTI-101 and peritoneal fluid and tumors were collected 10 days after treatment. Tumor-antigen specific T cells were stained using a CT26-specific tetramer antibody (MHC-H2Ld Gp70 SPSYVYHQF) and quantified using flow cytometry. Each dot represents an individual mouse. D, Flow cytometry was used to profile memory T cells on the same mice from C. E and F, Balb/c mice with established CT26-ip tumors (E) or C57Bl/6 mice with established B16F10-ip tumors (F) were treated with mSENTI-101 via intraperitoneal injection. Tumor burden was measured weekly by bioluminescence imaging of fLuciferase. Mice that were confirmed tumor-free more than 90 days after treatment with mSENTI-101 were re-challenged by subcutaneous implantation of parental tumor cells in the flank. A cohort of treatment-naïve mice were implanted with tumor cells in parallel to serve as control. Tumor volume was measured using a caliper. Mice were euthanized when tumor volume reached 1,500 mm3. Individual tumor growth curves are plotted for the CT26 (E) and B16F10 (F) tumors.

Close modal

Recent successes of immunotherapies in certain cancer indications have revolutionized the treatment paradigm in oncology. Unfortunately, most solid tumors, among them ovarian cancer and peritoneal carcinomatosis, remain unresponsive to these advances (13). Thus, there remains an unmet need for novel therapeutics that can effectively modify and activate the immune system to fight cancer.

Allogeneic BM-MSCs have been extensively used in the clinic, primarily for treatment of graft-versus-host diseases; however, the use of gene-engineered MSCs for the treatment of cancer is more limited (NCT02530047, NCT02008539; ref. 24). Despite remarkable success of engineered cell-based immune therapeutics, such as CAR-T cells that have revolutionized the treatment paradigm for liquid tumors, some important challenges remain. Autologous engineered cell-based therapeutics require a long and expensive manufacturing process. Furthermore, the immunosuppressive tumor microenvironment present in solid tumors is a barrier to efficacious treatments. MSCs offer several advantages over autologous T-cell therapeutics: (i) MSCs are naturally immune privileged, allowing for allogeneic off-the-shelf products; (ii) MSCs are easy to manufacture and have extensive expansion potential; (iii) MSCs innately localize and home into solid tumors, thus reducing the risk for systemic toxicities; and (iv) MSCs are not inhibited by the suppressive tumor microenvironment.

Here we present a novel cell-based therapeutic, SENTI-101, consisting of allogeneic MSCs as a modality to deliver two potent cytokines (IL12 and IL21) into the tumor microenvironment, to elicit durable immune-mediated antitumor responses and induce immune memory. The combination of IL12 and IL21 was chosen among more than 20 immune-stimulatory factors evaluated on the basis of in vivo antitumor efficacy. These two immunomodulatory cytokines (IL12 and IL21) engage both adaptive and innate immune responses against the tumor, have complementary and multimodal mechanisms of action, (39–41, 43, 44, 46) and have been shown to induce antitumor activity in multiple nonclinical models of cancer (40, 42, 43, 62). IL12 is a covalently linked heterodimer composed of two chains, p40 and p35, connected by a disulfide bridge, which is mainly secreted by subclasses of antigen-presenting cells (39). Its membrane receptor consists of two chains, IL12Rβ1 and IL12Rβ2, and is present on immune cell populations such as NK cells, CD4+ and CD8+ T cells. Upon binding to its receptor, IL12 promotes the secretion of Th1 cytokines (e.g., IFNγ) and the subsequent induction of an antitumor immune response. IL21 is a four-α-helical bundle cytokine mainly secreted by activated CD4+ T cells and iNKT cells. Its heterodimeric receptor (IL21R) comprises a distinct α-chain and a common cytokine receptor γ chain shared with the IL2 family of cytokines (46). IL21R has a pleiotropic function and is expressed on B cells, iNKT cells, CD4 and CD8 T cells as well as myeloid cells and is known to drive B-cell maturation and proliferation. In addition, IL21 can induce antitumor activities, from promoting NK function to supporting T-cell memory phenotype and persistence (44–46).

The use of cytokines for cancer immunotherapy has been extensively applied. For example, high-dose IL2 is approved for the treatment of metastatic renal cell carcinoma and metastatic melanoma (63). New approaches are under intense investigations, including late-stage clinical studies using pegylated IL2 (NKT-241) for the treatment of metastatic solid tumors (64, 65). Specifically, for IL12 and IL21, there are over 33 active clinical trials listed on Clinicaltrials.gov at the time of this writing. To date, two published examples of IL12-secreting cell therapies (lymphocytes) have been tested in clinical trials (NCT02498912; ref. 20, 66). A phase 1/2 study was conducted to evaluate the effect of IL12 expression under the control of a nuclear factor of activated T cells (NFAT) inducible promoter in autologous Tumor Infiltrating Lymphocytes (TILs) (20). This previous clinical experience with IL12-engineered T cells has shown that IL12-secreting cell therapy is feasible. However, the success of these strategies as well as previous clinical experiences using recombinant cytokines (17) has been hampered by severe toxicities due to systemic and uncontrolled immune activation, as well as the lack of efficacy caused by the poor PKs and short half-lives of these compounds. Recently, GEN-1, an IL12-encoding plasmid has been used with remarkable success in combination with neoadjuvant chemotherapy in early-stage clinical trials for ovarian cancer (NCT02480374). By leveraging the tumor-homing capability of MSCs to locally produce IL12 and IL21 in the tumor environment, the risk of systemic exposure may be significantly reduced, while broadening the effective therapeutic window of these cytokines in the locoregional tumor area.

Although MSCs innate function is to suppress immune activation and have been involved in cancer initiation or promotion (67), the expression of two potent immune stimulatory cytokines, IL12 and IL21, results in an overall antitumor immune response. In all our preclinical studies, control MSCs (engineered with an empty vector) did not demonstrate tumor increase or acceleration, which could be explained by the fact that we treated established (∼100 mg) tumors in syngeneic hosts. The preclinical and mechanistic studies described here demonstrate that SENTI-101 is able to act on multiple steps of the cancer immunity cycle (58), engaging both innate and adaptive immune compartments. Mice treated with SENTI-101 showed significant increase in antigen-presenting cells in their lymph nodes, characterized by expression of markers CD103 and XCR1. These cells have been demonstrated to enable the differentiation of T-cell memory phenotypes and a subsequent sustained antitumor response despite tumor or pathogen rechallenge (58, 60, 61). By activating both the innate and adaptive immune responses, SENTI-101 increases dendritic cells in the tumor-draining lymph nodes, T-cell local activation in the peritoneal fluid, and results in increased tumor-specific T cells and T-cell memory subsets. Our mechanistic studies using immunocompetent tumor models corroborate that this multimodal engagement of the cancer immunity cycle results in durable antitumor responses and immune memory as evidenced by rejection of newly implanted tumors in mice that were cured after treatment with SENTI-101.

SENTI-101 outperformed the efficacy and response rates of checkpoint inhibitor anti-PD1 in both preclinical models. The proposed mechanism of action of SENTI-101, with increased abundance and expression of activation markers on T cells, B cells, and APCs, would be indicative of turning “cold” tumors hot.” Our results suggest that Senti-101 could synergize or improve the response rate in those cases in which checkpoint inhibitors are not efficacious, as is the case of ovarian cancer. Analysis of clinical samples from HGSOC reveals the importance of the tumor immune infiltrate as a prognostic factor. Immune correlates induced by SENTI-101 such as the chemokines CXCL9 and 10, IFNγ, dendritic cells, T cells, B cells, and tertiary lymphoid structures (TLS) have been correlated with better prognosis in patients with ovarian cancer (55, 68–70). Recently, multiple studies have shown the importance of the immune landscape and the presence of T cells and B cells organized in TLS in determining response to checkpoint inhibitors (51–54), further supporting the testing of SENTI-101 in solid tumors either as a single agent or in combination with checkpoint inhibitors. Importantly, SENTI-101 demonstrated antitumor activity in different solid tumor models with very distinct immune landscapes such as CT26 (immune rich or “hot”) and B16F10 (immune excluded or “cold”), which suggests a broad applicability for SENTI-101 in multiple solid tumors, where cytokines could be utilized to polarize and re-educate the tumor microenvironment resulting in robust immune-mediated tumor-cell killing.

A. GonzalezJunca reports other support from Senti Biosciences, Inc., during the conduct of the study; also has a patent for US10993967 issued and a patent for US20200206271 pending. F.D. Liu reports other support from Senti Biosciences, Inc. during the conduct of the study. A. Mullenix reports other support from Senti Biosciences, Inc. during the conduct of the study. C. Lee reports other support from Senti B Biosciences, Inc. during the conduct of the study. R.M. Gordley reports other support from Senti Biosciences, Inc., during the conduct of the study; also has a patent for US10993967 issued and a patent for US20200206271 pending. D.O. Frimannsson reports other support from Senti Biosciences, Inc. during the conduct of the study; also has a patent for US Patent Application Publication No. US20200206271 pending to SENTI BIOSCIENCE and US Patent No. US10993967 issued to SENTI BIOSCIENCE. O. Maller reports other support from Senti Biosciences, Inc. during the conduct of the study; other support from Adicet Bio outside the submitted work. D. Iyer reports other support from Senti Biosciences, Inc. during the conduct of the study. M. Tian reports other support from Biosciences, Inc. during the conduct of the study. R. Martinez reports other support from Senti Biosciences, Inc. during the conduct of the study. R. Savur reports other support from Senti Biosciences, Inc. during the conduct of the study. A. Perry-McNamara reports personal fees from Senti Biosciences, Inc. during the conduct of the study. D. Nguyen reports other support from Senti Biosciences, Inc. during the conduct of the study. N. Almudhfar reports other support from Senti Biosciences, Inc. during the conduct of the study. C. Blanco reports other support from Senti Biosciences, Inc. during the conduct of the study. C. Huynh reports other support from Senti Biosciences, Inc. during the conduct of the study. A. Nand reports other support from Senti Bioscience, Inc. during the conduct of the study. A. Magal reports other support from Senti Biosciences, Inc. during the conduct of the study. S. Mangalampalli reports other support from Senti Biosciences, Inc. during the conduct of the study. P.J. Lee reports other support from Biosciences, Inc. outside the submitted work; also has a patent for US20200206271 pending to Senti Biosciences, Inc. and US10993967 issued to Biosciences, Inc. T.K. Lu reports other support from Senti Biosciences, Inc. during the conduct of the study; also has a patent for US Patent Application Publication No. US20200206271 pending to Senti Biosciences, Inc. and a US Patent No. US10993967 issued to Senti Biosciences, Inc; and is a co-founder of Senti Biosciences Inc., Synlogic, Engine Biosciences, Tango Therapeutics, Corvium, BiomX, Eligo Biosciences, Bota.Bio, Avendesora, and NE47Bio; he also holds financial interests in nest.bio, Armata, IndieBio, CognitoHealth, Quark Biosciences, Personal Genomics, Thryve, Lexent Bio, MitoLab, Vulcan, Serotiny, Avendesora, Pulmobiotics, Provectus Algae, Invaio, NSG Biolabs. G. Lee reports other support from Senti Biosciences outside the submitted work. No disclosures were reported by the other authors.

A. Gonzalez-Junca: Conceptualization, formal analysis, supervision, investigation, methodology, writing–original draft, writing–review and editing. F.D. Liu: Conceptualization, formal analysis, investigation, methodology, writing–original draft. A.S. Nagaraja: Conceptualization, formal analysis, investigation, methodology. A. Mullenix: Formal analysis, investigation, methodology. C-T. Lee: Supervision, investigation, methodology. R.M. Gordley: Conceptualization, resources, supervision. D.O. Frimannsson: Conceptualization, resources. O. Maller: Conceptualization, methodology. B.S. Garrison: Conceptualization, methodology. D. Iyer: Supervision, methodology. A. Benabbas: Resources, methodology. T.A. Truong: Methodology. A. Quach: Methodology. M. Tian: Methodology. R. Martinez: Methodology. R. Savur: Data curation, methodology. A. Perry-McNamara: Methodology. D. Nguyen: Methodology. N. Almudhfar: Methodology. C. Blanco: Resources, methodology. C. Huynh: Resources. A. Nand: Resources. Y.-A. E. Lay: Resources, methodology. A. Magal: Resources, methodology. S. Mangalampalli: Resources, methodology. P.J. Lee: Conceptualization, supervision, funding acquisition. T.K. Lu: Conceptualization, supervision, funding acquisition. G. Lee: Conceptualization, supervision, funding acquisition, project administration, writing–review and editing.

1.
Coccolini
F
. 
Peritoneal carcinomatosis
.
World J Gastroenterol
2013
;
19
:
6979
.
2.
Lengyel
E
. 
Ovarian cancer development and metastasis
.
Am J Pathol
2010
;
177
:
1053
64
.
3.
Weidle
UH
,
Birzele
F
,
Kollmorgen
G
,
Rueger
R
. 
Mechanisms and targets involved in dissemination of ovarian cancer
.
Cancer Genomics Proteomics
2016
;
13
:
407
24
.
4.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2019
.
CA Cancer J Clin
. 
2019
;
69
:
7
34
.
5.
van Driel
WJ
,
Koole
SN
,
Sikorska
K
,
Schagen van Leeuwen
JH
,
Schreuder
HWR
,
Hermans
RHM
, et al
Hyperthermic intraperitoneal chemotherapy in ovarian cancer
.
N Engl J Med
2018
;
378
:
230
40
.
6.
Tewari
D
,
Java
JJ
,
Salani
R
,
Armstrong
DK
,
Markman
M
,
Herzog
T
, et al
Long-term survival advantage and prognostic factors associated with intraperitoneal chemotherapy treatment in advanced ovarian cancer: a Gynecologic Oncology Group Study
.
J Clin Oncol
2015
;
33
:
1460
6
.
7.
Jiang
X
,
Li
W
,
Li
X
,
Bai
H
,
Zhang
Z
. 
Current status and future prospects of PARP inhibitor clinical trials in ovarian cancer
.
Cancer Manag Res
2019
;
11
:
4371
90
.
8.
Moore
K
,
Colombo
N
,
Scambia
G
,
Kim
B-G
,
Oaknin
A
,
Friedlander
M
, et al
Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer
.
N Engl J Med
2018
;
379
:
2495
505
.
9.
Raja
FA
,
Counsell
N
,
Colombo
N
,
Pfisterer
J
,
du Bois
A
,
Parmar
MK
, et al
Platinum versus platinum-combination chemotherapy in platinum-sensitive recurrent ovarian cancer: a meta-analysis using individual patient data
.
Ann Oncol
2013
;
24
:
3028
34
.
10.
Pujade-Lauraine
E
,
Hilpert
F
,
Weber
B
,
Reuss
A
,
Poveda
A
,
Kristensen
G
, et al
Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial
.
J Clin Oncol
2014
;
32
:
1302
8
.
11.
Ribas
A
,
Wolchok
JD
. 
Cancer immunotherapy using checkpoint blockade
.
Science
2018
;
359
:
1350
5
.
12.
Borella
F
,
Ghisoni
E
,
Giannone
G
,
Cosma
S
,
Benedetto
C
,
Valabrega
G
, et al
Immune checkpoint inhibitors in epithelial ovarian cancer: an overview on efficacy and future perspectives
.
Diagnostics
2020
;
10
:
146
.
13.
Matulonis
UA
,
Shapira-Frommer
R
,
Santin
AD
,
Lisyanskaya
AS
,
Pignata
S
,
Vergote
I
, et al
Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study
.
Ann Oncol
2019
;
30
:
1080
7
.
14.
Disis
ML
,
Taylor
MH
,
Kelly
K
,
Beck
JT
,
Gordon
M
,
Moore
KM
, et al
Efficacy and safety of avelumab for patients with recurrent or refractory ovarian cancer: phase 1b results from the JAVELIN solid tumor trial
.
JAMA Oncol
2019
;
5
:
393
.
15.
Hamanishi
J
,
Mandai
M
,
Ikeda
T
,
Minami
M
,
Kawaguchi
A
,
Murayama
T
, et al
Safety and antitumor activity of anti–PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer
.
J Clin Oncol
2015
;
33
:
4015
22
.
16.
Anwer
K
,
Barnes
MN
,
Fewell
J
,
Lewis
DH
,
Alvarez
RD
. 
Phase-I clinical trial of IL-12 plasmid/lipopolymer complexes for the treatment of recurrent ovarian cancer
.
Gene Ther
2010
;
17
:
360
9
.
17.
Lenzi
R
,
Edwards
R
,
June
C
,
Seiden
MV
,
Garcia
ME
,
Rosenblum
M
, et al
Phase II study of intraperitoneal recombinant interleukin-12 (rhIL-12) in patients with peritoneal carcinomatosis (residual disease < 1 cm) associated with ovarian cancer or primary peritoneal carcinoma
.
J Transl Med
2007
;
5
:
66
.
18.
Cohen
CA
,
Shea
AA
,
Heffron
CL
,
Schmelz
EM
,
Roberts
PC
. 
Interleukin-12 immunomodulation delays the onset of lethal peritoneal disease of ovarian cancer
.
J Interferon Cytokine Res
2016
;
36
:
62
73
.
19.
Strauss
J
,
Heery
CR
,
Kim
JW
,
Jochems
C
,
Donahue
RN
,
Montgomery
AS
, et al
First-in-human phase I trial of a tumor-targeted cytokine (NHS-IL12) in subjects with metastatic solid tumors
.
Clin Cancer Res
2019
;
25
:
99
109
.
20.
Zhang
L
,
Morgan
RA
,
Beane
JD
,
Zheng
Z
,
Dudley
ME
,
Kassim
SH
, et al
Tumor-infiltrating lymphocytes genetically engineered with an inducible gene encoding interleukin-12 for the immunotherapy of metastatic melanoma
.
Clin Cancer Res
2015
;
21
:
2278
88
.
21.
Conlon
KC
,
Miljkovic
MD
,
Waldmann
TA
. 
Cytokines in the treatment of cancer
.
J Interferon Cytokine Res
2019
;
39
:
6
21
.
22.
Berraondo
P
,
Sanmamed
MF
,
Ochoa
MC
,
Etxeberria
I
,
Aznar
MA
,
Pérez-Gracia
JL
, et al
Cytokines in clinical cancer immunotherapy
.
Br J Cancer
2019
;
120
:
6
15
.
23.
Galipeau
J
,
Sensébé
L
. 
Mesenchymal stromal cells: clinical challenges and therapeutic opportunities
.
Cell Stem Cell
2018
;
22
:
824
33
.
24.
Andreeff
M
,
Marini
FC
,
Westin
SN
,
Coleman
RL
,
Thall
PF
,
Aljahdami
V
, et al
Abstract 75: a phase I trial of mesenchymal stem cells transfected with a plasmid secreting interferon beta in advanced ovarian cancer
.
Tumor Biol.
2018
:
78
:
AM2018-75
.
25.
Viswanathan
S
,
Shi
Y
,
Galipeau
J
,
Krampera
M
,
Leblanc
K
,
Martin
I
, et al
Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy (ISCT®) Mesenchymal Stromal Cell committee position statement on nomenclature
.
Cytotherapy
2019
;
21
:
1019
24
.
26.
Pittenger
MF
,
Discher
DE
,
Péault
BM
,
Phinney
DG
,
Hare
JM
,
Caplan
AI
. 
Mesenchymal stem cell perspective: cell biology to clinical progress
.
NPJ Regen Med
2019
;
4
:
22
.
27.
Gao
F
,
Chiu
SM
,
Motan
DAL
,
Zhang
Z
,
Chen
L
,
Ji
H-L
, et al
Mesenchymal stem cells and immunomodulation: current status and future prospects
.
Cell Death Dis
2016
;
7
:
e2062
.
28.
Zhao
Q
,
Ren
H
,
Han
Z
. 
Mesenchymal stem cells: Immunomodulatory capability and clinical potential in immune diseases
.
J Cell Immunother
2016
;
2
:
3
20
.
29.
Fitzsimmons
REB
,
Mazurek
MS
,
Soos
A
,
Simmons
CA
. 
Mesenchymal stromal/stem cells in regenerative medicine and tissue engineering
.
Stem Cells Int
2018
;
2018
:
1
16
.
30.
Rawat
S
,
Gupta
S
,
Mohanty
S
. 
Mesenchymal stem cells modulate the immune system in developing therapeutic interventions
.
In:
Tyagi
R
,
Bisen
PS
,
editors.
Immune response activation and immunomodulation
.
IntechOpen
; 
2019
[cited 2020 Aug 30].
https://www.intechopen.com/books/immune-response-activation-and-immunomodulation/mesenchymal-stem-cells-modulate-the-immune-system-in-developing-therapeutic-interventions.
31.
Kidd
S
,
Spaeth
E
,
Dembinski
JL
,
Dietrich
M
,
Watson
K
,
Klopp
A
, et al
Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging
.
Stem Cells
2009
;
27
:
2614
23
.
32.
Ullah
M
,
Liu
DD
,
Thakor
AS
. 
Mesenchymal stromal cell homing: mechanisms and strategies for improvement
.
iScience.
2019
;
15
:
421
38
.
33.
Kyburz
KA
,
Anseth
KS
. 
Three-dimensional hMSC motility within peptide-functionalized PEG-based hydrogels of varying adhesivity and crosslinking density
.
Acta Biomater
2013
;
9
:
6381
92
.
34.
Khalil
C
,
Moussa
M
,
Azar
A
,
Tawk
J
,
Habbouche
J
,
Salameh
R
, et al
Anti-proliferative effects of mesenchymal stem cells (MSCs) derived from multiple sources on ovarian cancer cell lines: an in-vitro experimental study
.
J Ovarian Res
2019
;
12
:
70
.
35.
Almeida-Porada
G
,
Atala
AJ
,
Porada
CD
. 
Therapeutic mesenchymal stromal cells for immunotherapy and for gene and drug delivery
.
Mol Ther Methods Clin Dev
2020
;
16
:
204
24
.
36.
Studeny
M
,
Marini
FC
,
Champlin
RE
,
Zompetta
C
,
Fidler
IJ
,
Andreeff
M
. 
Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors
.
Cancer Res
2002
;
62
:
3603
8
.
37.
Yu
JW
,
Bhattacharya
S
,
Yanamandra
N
,
Kilian
D
,
Shi
H
,
Yadavilli
S
, et al
Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironments
.
PLoS One
2018
;
13
:
e0206223
.
38.
Mosely
SIS
,
Prime
JE
,
Sainson
RCA
,
Koopmann
J-O
,
Wang
DYQ
,
Greenawalt
DM
, et al
Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery
.
Cancer Immunol Res
2017
;
5
:
29
41
.
39.
Trinchieri
G
,
Pflanz
S
,
Kastelein
RA
. 
The IL-12 family of heterodimeric cytokines
.
Immunity
2003
;
19
:
641
4
.
40.
Lasek
W
,
Zagożdżon
R
,
Jakobisiak
M
. 
Interleukin 12: still a promising candidate for tumor immunotherapy?
Cancer Immunol Immunother
2014
;
63
:
419
35
.
41.
Zundler
S
,
Neurath
MF
. 
Interleukin-12: functional activities and implications for disease
.
Cytokine Growth Factor Rev
2015
;
26
:
559
68
.
42.
Yeku
OO
,
Purdon
TJ
,
Koneru
M
,
Spriggs
D
,
Brentjens
RJ
. 
Armored CAR T cells enhance antitumor efficacy and overcome the tumor microenvironment
.
Sci Rep
2017
;
7
:
10541
.
43.
Croce
M
,
Rigo
V
,
Ferrini
S
. 
IL-21: a pleiotropic cytokine with potential applications in oncology
.
J Immunol Res
2015
;
2015
:
1
15
.
44.
Skak
K
,
Frederiksen
KS
,
Lundsgaard
D
. 
Interleukin-21 activates human natural killer cells and modulates their surface receptor expression
.
Immunology
2008
;
123
:
575
83
.
45.
Kuchen
S
,
Robbins
R
,
Sims
GP
,
Sheng
C
,
Phillips
TM
,
Lipsky
PE
, et al
Essential role of IL-21 in B cell activation, expansion, and plasma cell generation during CD4 + T Cell-B cell collaboration
.
J Immunol
2007
;
179
:
5886
96
.
46.
Davis
MR
,
Zhu
Z
,
Hansen
DM
,
Bai
Q
,
Fang
Y
. 
The role of IL-21 in immunity and cancer
.
Cancer Lett
2015
;
358
:
107
14
.
47.
Frimannsson
D
,
Garrison
BS
,
Gonzalez-Junca
A
,
Gordley
RM
,
Lee
PJ
, et al
“Combinatorial Cancer Immunotherapy” U.S. patent application number 16/656452 was filed with the patent office on 2020-06-04
. https://patents.google.com/patent/US20200171093A1/en.
48.
Watanabe
H
,
Numata
K
,
Ito
T
,
Takagi
K
,
Matsukawa
A
. 
Innate immune response in Th1- and Th2-dominant mouse strains
.
Shock
2004
;
22
:
460
6
.
49.
Castro
F
,
Cardoso
AP
,
Gonçalves
RM
,
Serre
K
,
Oliveira
MJ
. 
Interferon-gamma at the crossroads of tumor immune surveillance or evasion
.
Front Immunol
2018
;
9
:
847
.
50.
Miller
G
,
Bleier
JI
,
Antonescu
C
,
Pillarisetty
VG
,
Shah
AB
,
Lahrs
S
, et al
Natural killer cell depletion confounds the antitumor mechanism of endogenous IL-12 overexpression
.
Int J Cancer
2004
;
110
:
395
402
.
51.
Sautès-Fridman
C
,
Petitprez
F
,
Calderaro
J
,
Fridman
WH
. 
Tertiary lymphoid structures in the era of cancer immunotherapy
.
Nat Rev Cancer
2019
;
19
:
307
25
.
52.
Cabrita
R
,
Lauss
M
,
Sanna
A
,
Donia
M
,
Larsen
MS
,
Mitra
S
, et al
Tertiary lymphoid structures improve immunotherapy and survival in melanoma
.
Nature
2020
;
577
:
561
5
.
53.
Petitprez
F
,
de Reyniès
A
,
Keung
EZ
,
Chen
TW-W
,
Sun
C-M
,
Calderaro
J
, et al
B cells are associated with survival and immunotherapy response in sarcoma
.
Nature
2020
;
577
:
556
60
.
54.
Helmink
BA
,
Reddy
SM
,
Gao
J
,
Zhang
S
,
Basar
R
,
Thakur
R
, et al
B cells and tertiary lymphoid structures promote immunotherapy response
.
Nature
2020
;
577
:
549
55
.
55.
Nielsen
JS
,
Sahota
RA
,
Milne
K
,
Kost
SE
,
Nesslinger
NJ
,
Watson
PH
, et al
CD20+ tumor-infiltrating lymphocytes have an atypical CD27- memory phenotype and together with CD8+ T cells promote favorable prognosis in ovarian cancer
.
Clin Cancer Res
2012
;
18
:
3281
92
.
56.
Parisi
G
,
Saco
JD
,
Salazar
FB
,
Tsoi
J
,
Krystofinski
P
,
Puig-Saus
C
, et al
Persistence of adoptively transferred T cells with a kinetically engineered IL-2 receptor agonist
.
Nat Commun
2020
;
11
:
660
.
57.
Xue
Q
,
Bettini
E
,
Paczkowski
P
,
Ng
C
,
Kaiser
A
,
McConnell
T
, et al
Single-cell multiplexed cytokine profiling of CD19 CAR-T cells reveals a diverse landscape of polyfunctional antigen-specific response
.
J Immunother Cancer
2017
;
5
:
85
.
58.
Chen
DS
,
Mellman
I
. 
Oncology meets immunology: the cancer-immunity cycle
.
Immunity
2013
;
39
:
1
10
.
59.
Chen
DS
,
Mellman
I
. 
Elements of cancer immunity and the cancer–immune set point
.
Nature
2017
;
541
:
321
30
.
60.
Broz
ML
,
Binnewies
M
,
Boldajipour
B
,
Nelson
AE
,
Pollack
JL
,
Erle
DJ
, et al
Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity
.
Cancer Cell
2014
;
26
:
638
52
.
61.
Binnewies
M
,
Mujal
AM
,
Pollack
JL
,
Combes
AJ
,
Hardison
EA
,
Barry
KC
, et al
Unleashing Type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity
.
Cell.
2019
;
177
:
556
71
.e16.
62.
Koneru
M
,
Purdon
TJ
,
Spriggs
D
,
Koneru
S
,
Brentjens
RJ
. 
IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors in vivo
.
OncoImmunology
2015
;
4
:
e994446
.
63.
Jiang
T
,
Zhou
C
,
Ren
S
. 
Role of IL-2 in cancer immunotherapy
.
OncoImmunology
2016
;
5
:
e1163462
.
64.
Bentebibel
S-E
,
Hurwitz
ME
,
Bernatchez
C
,
Haymaker
C
,
Hudgens
CW
,
Kluger
HM
, et al
A First-in-human study and biomarker analysis of NKTR-214, a Novel IL2Rβγ-biased cytokine, in patients with advanced or metastatic solid tumors
.
Cancer Discov
2019
;
9
:
711
21
.
65.
Diab
A
,
Tannir
NM
,
Bentebibel
S-E
,
Hwu
P
,
Papadimitrakopoulou
V
,
Haymaker
C
, et al
Bempegaldesleukin (NKTR-214) plus nivolumab in patients with advanced solid tumors: phase i dose-escalation study of safety, efficacy, and immune activation (PIVOT-02)
.
Cancer Discov
2020
;
10
:
1158
73
.
66.
Koneru
M
,
O'Cearbhaill
R
,
Pendharkar
S
,
Spriggs
DR
,
Brentjens
RJ
. 
A phase I clinical trial of adoptive T cell therapy using IL-12 secreting MUC-16ecto directed chimeric antigen receptors for recurrent ovarian cancer
.
J Transl Med
2015
;
13
:
102
.
67.
Sai
B
,
Dai
Y
,
Fan
S
,
Wang
F
,
Wang
L
,
Li
Z
, et al
Cancer-educated mesenchymal stem cells promote the survival of cancer cells at primary and distant metastatic sites via the expansion of bone marrow-derived-PMN-MDSCs
.
Cell Death Dis
2019
;
10
:
941
.
68.
Truxova
I
,
Kasikova
L
,
Hensler
M
,
Skapa
P
,
Laco
J
,
Pecen
L
, et al
Mature dendritic cells correlate with favorable immune infiltrate and improved prognosis in ovarian carcinoma patients
.
J Immunother cancer
2018
;
6
:
139
.
69.
Bronger
H
,
Singer
J
,
Windmüller
C
,
Reuning
U
,
Zech
D
,
Delbridge
C
, et al
CXCL9 and CXCL10 predict survival and are regulated by cyclooxygenase inhibition in advanced serous ovarian cancer
.
Br J Cancer
2016
;
115
:
553
63
.
70.
Zhang
L
,
Conejo-Garcia
JR
,
Katsaros
D
,
Gimotty
PA
,
Massobrio
M
,
Regnani
G
, et al
Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer
.
N Engl J Med
2003
;
348
:
203
13
.

Supplementary data