Purpose:

To explore the cellular cross-talk of tumor-resident mast cells (MC) in controlling the activity of cancer-associated fibroblasts (CAF) to overcome tumor microenvironment (TME) abnormalities, enhancing the efficacy of immune-checkpoint inhibitors in sarcoma.

Experimental Design:

We used a coculture system followed by further validation in mouse models of fibrosarcoma and osteosarcoma with or without administration of the MC stabilizer and antihistamine ketotifen. To evaluate the contribution of ketotifen in sensitizing tumors to therapy, we performed combination studies with doxorubicin chemotherapy and anti–PD-L1 (B7-H1, clone 10F.9G2) treatment. We investigated the ability of ketotifen to modulate the TME in human sarcomas in the context of a repurposed phase II clinical trial.

Results:

Inhibition of MC activation with ketotifen successfully suppressed CAF proliferation and stiffness of the extracellular matrix accompanied by an increase in vessel perfusion in fibrosarcoma and osteosarcoma as indicated by ultrasound shear wave elastography imaging. The improved tissue oxygenation increased the efficacy of chemoimmunotherapy, supported by enhanced T-cell infiltration and acquisition of tumor antigen–specific memory. Importantly, the effect of ketotifen in reducing tumor stiffness was further validated in sarcoma patients, highlighting its translational potential.

Conclusions:

Our study suggests the targeting of MCs with clinically administered drugs, such as antihistamines, as a promising approach to overcome resistance to immunotherapy in sarcomas.

Translational Relevance

Immune-checkpoint inhibitors (ICI) have been a breakthrough in cancer treatment, but not all patients respond favorably, including sarcoma patients. The efficacy of ICIs is impeded by abnormalities in the tumor microenvironment (TME), which foster immunosuppression. In this study, we demonstrate that the antihistamine drug ketotifen not only suppresses extracellular matrix formation but also enhances anti–PD-L1 immunotherapy by reversing the immunosuppressive TME, leading to increased overall survival in preclinical models of fibrosarcoma and osteosarcoma. These therapeutic effects align closely with a reduction in tumor stiffness and an increase in vascular perfusion, suggesting that TME priming with ketotifen is a prerequisite to creating favorable immunogenic conditions capable of eliminating the entirety of the tumor. Furthermore, within the context of a repurposed phase II clinical trial, we provide the first-time evidence of ketotifen's ability to modulate the TME in human sarcomas, offering a compelling rationale for clinical translation.

Harnessing the immune system to fight cancer has led to significant clinical benefits in many tumor types. The most successful example of cancer immunotherapy is the use of immune-checkpoint inhibitors (ICI). However, ICI responses are limited to patients whose tumors are infiltrated by a sufficiently diverse repertoire of cytotoxic CD8+ T cells (1, 2). Apart from lymphocyte infiltration, other features of the tumor microenvironment (TME) are likely to obstruct antitumor immune responses and lead to treatment failure in sarcomas (3, 4). In fact, cancer-associated fibroblasts (CAF), which is the most common nonmalignant cell type in many aggressive solid tumors, can produce and maintain elevated levels of extracellular matrix (ECM) components, including collagen I and hyaluronan. Accumulation of these structural components causes the stiffening of the tumor and abnormal elevation of mechanical forces that compress tumor vessels, leading to their collapse (5–9). Notably, in tumors with an abundance of ECM, 95% of intratumoral blood vessels might be compressed and up to 80% are totally collapsed and lacking perfusion (10). Hypoperfusion, in turn, can induce hypoxia, resulting in increased immunosuppressive signaling, which hampers antitumor immunity (11–13). Many types of sarcomas are typical examples of these TME abnormalities, whereas restoring TME function to normal levels (i.e., TME normalization) has been proven to be an effective strategy to potentiate immunotherapy in stroma-rich tumors, such as breast and pancreatic cancers (10, 14–16). The effect of normalization strategies in sarcoma tumors has not been studied yet (3).

Indeed, current TME normalization strategies utilizing drugs that directly reprogram CAFs, such as TGFβ inhibitors, can overcome the ECM barrier to immune cell infiltration and enhance the delivery of ICIs (14, 15, 17–21). Nevertheless, resistance to immunotherapy is given not only by the inability of immunotherapy to reach and eliminate cancer cells but also by the immunosuppressive microenvironment which dampens the activation of cytotoxic T cells and prevents immunotherapy from establishing a lasting immune protection against tumor recurrence. This challenge calls for a sophisticated harnessing of immune cells, such as the tumor-resident mast cells (MC), whose biological function can be programmed. MCs infiltrate the TME with a not yet fully developed or matured phenotype that can be influenced by the surrounding milieu (22) to play a protumorigenic (23, 24) or an antitumorigenic role (25), which can involve interaction with CAFs (26). Upon activation, MCs release secretory granules containing mediators that influence adaptive immune responses, ECM remodeling, and angiogenesis (27–31). Increasing evidence suggests that human MCs release VEGF-A and IL6 in response to hypoxia (32, 33), which favors immunosuppression and poor response to immunotherapy (34–36). Thus, targeting MCs with clinically administered drugs such as antihistamines, MC stabilizers (e.g., ketotifen, cromolyn), or other MC-targeting drugs represents a promising approach to overcome resistance to current cancer treatment modalities (37–39). The functional plasticity of MCs makes them key orchestrators of the immune TME, and their inhibition may hold the key to breaking tumor resistance to immunotherapy.

In this study, we aimed to explore the role of tumor-resident MCs in modulating antitumor responses either as a core component of the host immune system or via their interaction with CAFs. MC–fibroblast interactions are well characterized in the context of wound healing and pulmonary fibrosis. In these settings, MCs are in close proximity to fibroblasts acting as potent regulators of fibroblast proliferation and collagen synthesis (40, 41). Nonetheless, their dynamic interaction with CAFs in tumors is not well studied. Understanding the dynamic cross-talk between MCs and CAFs could lead to new therapeutic strategies that reprogram MCs and CAFs to quiescent phenotypes, reducing levels of ECM, angiogenesis, and immunosuppression.

Here, we hypothesize that reprogramming of MCs can normalize tumor stiffness and perfusion in murine sarcoma models through their interaction with CAFs. Specifically, we investigate the effects of ketotifen, a second-generation histamine H1 blocker and mast cell stabilizer, on modulating the TME. We demonstrate that ketotifen can reprogram MCs toward effectively suppressing CAF-induced stiffness elevation, leading to increased perfusion in murine models of soft-tissue and bone sarcoma. Through this unexplored mechanism of MC and CAF reprogramming, we show that ketotifen's combination with chemotherapy and PD-L1 blockade overcomes the primary resistance of these tumor models to immunotherapy, resulting in extended survival and immunologic memory. Furthermore, as part of a phase II clinical trial (EU clinical trials register, EudraCT #: 2022-002311-39), we show that administration of ketotifen in patients with sarcoma recapitulates the reduction in tumor stiffness observed in the tumor mouse models for different sarcoma subtypes.

Cell lines and cell culture

The MCA205 mouse fibrosarcoma cell line was purchased from Millipore (SCC173, Millipore) and cultured in expansion medium consisting of Roswell Park Memorial Institute medium (RPMI-1640, LM-R1637, Biosera) containing 2 mmol/L L-glutamine (TMS-002-C, Sigma), 1 mmol/L sodium pyruvate (TMS-005-C, Sigma), 10% fetal bovine serum (FBS, FB-1001H, Biosera), 1× nonessential amino acids (TMS-001-C, Sigma), 1% antibiotics (A5955, Sigma), and 1× β-mercaptoethanol (ES-007-E, Sigma). The K7M2 mouse osteosarcoma cell line (CRL283) and NIH3T3 fibroblast cell line (CRL-1658) were obtained from ATCC and cultured in Dulbecco's Modified Eagle's Medium (DMEM, LM-S2041, Biosera) supplemented with 10% FBS and 1% antibiotics. The MC/9 mast cell line was purchased from ATCC (CRL-830, ATCC) and cultured in RPMI medium supplemented with 10% FBS, 1% antibiotics, 1× β-mercaptoethanol, 100 ng/mL stem cell factor (SCF, 250–03, PeproTech), and 10 ng/mL IL3 (213–13, PeproTech). All cells were maintained in humidified incubators at 37°C with 5% CO2 and routinely tested and confirmed to be free of mycoplasma contamination.

CAFs were isolated from MCA205 fibrosarcoma tumors with a size of 200 mm3. Briefly, resected tumors were washed in 1× PBS, finely minced, and incubated in Liberase solution for 30 minutes at 37°C. Enzymatic activity was ceased with a complete culture medium and cell suspension was passed through a 40-μm cell strainer to obtain single cells. Filtrate was centrifuged at 300 × g at 4°C for 5 minutes, the supernatant was removed completely, and the cell pellet was resuspended in a complete growth medium and transferred into a cell culture vessel at 37°C/5% CO2. The cells attaching to the vessel bottom within 20 minutes were recognized as CAFs, and the floating cells were abandoned. This isolation process was repeated 3 times to get purified CAFs.

Drugs and reagents

Ketotifen fumarate salt (K2628, Sigma) was dissolved in sterile normal saline (0.9% NaCl in ddH2O, w/v). Mouse monoclonal antibody anti–PD-L1 (B7-H1, clone 10F.9G2, RRID: AB_10949073) and rat IgG2b isotype control, antikeyhole limpet hemocyanin (LTF-2, RRID: AB_1107780) were purchased from Bio X Cell and dissolved in the recommended InVivoPure pH 7.0 Dilution Buffer (IP0070). Doxorubicin hydrochloride was obtained from Nicosia General Hospital as a ready-made solution of 2 mg/mL. Anti–PD-L1 was administered via intraperitoneal injection at a dose of 10 mg/kg (42) and doxorubicin at a dose of 5 mg/kg (14).

Animal care and mouse models

Six-week-old C57BL/6OlaHsd (RRID:MGI:5656288) female mice were injected in the leg muscle with 2.5×105 MCA205 fibrosarcoma cells. The osteosarcoma syngeneic tumor model was generated by orthotopic implantation of 1×106 wild-type K7M2 cells in the proximal tibia of 6-week-old BALB/cOlaHsd (RRID:MGI:5653315) female mice as previously described (43). All mice were maintained in specific pathogen-free conditions in the animal facilities of the Cyprus Institute of Neurology and Genetics. They were housed in a controlled temperature/humidity (22°C/55%) environment on a 12-hour light–dark cycle and kept with free access to food and water throughout the whole experiment period. A sample size identical to that of previous studies, which measured the same output variables, was used. All in vivo experiments were conducted per the animal welfare regulations and guidelines of the European Union (European Directive 2010/63/EE and Cyprus Legislation for the protection and welfare of animals, Laws 1994–2013) under a license acquired and approved (CY/EXP/PR.L2/2018, CY/EXP/PR.L14/2019, CY/EXP/PR.L15/2019, and CY/EXP/PR.L03/2020) by the Cyprus Veterinary Services Committee, the Cyprus national authority for monitoring the welfare of animals in research.

Ketotifen dose-response studies

Ketotifen at 1, 5, 10, and 25 mg/kg or an equal volume of diluent (control group) was administered daily by intraperitoneal injection (i.p.) for 8 days. Ketotifen treatment for MCA205 and K7M2 tumor models started when the average tumor volume reached 60 mm3 and 150 mm3, respectively, and was completed at 600 mm3. At the study endpoint, primary tumors were surgically excised and stored in 1× PBS at −80°C until further processing for immunofluorescence staining experiments.

Ketotifen combination with chemoimmunotherapy

Mice bearing MCA205 tumors were pretreated with daily ketotifen at the dose of 10 mg/kg (found to be optimal in this study) or an equal volume of diluent (control group) before the chemoimmunotherapy combination treatment. Specifically, ketotifen treatment commenced when the average volume of MCA205 tumors was 40 mm3. The first cycle of doxorubicin and anti–PD-L1 combination occurred when tumors reached an average size of 150 mm3 (day 7) and repeated for an additional two cycles with a three-day interval (i.e., on days 10 and 13 after cancer cell implantation). Ketotifen administration continued for an additional two days after the last cycle of combination treatment. Following completion of the study protocol, primary tumors having an average size of 700 mm3 (on day 16) were resected and processed accordingly for the flow cytometry experiment, and survival of mice was monitored daily until the rechallenge experiment.

In the K7M2 tumor model, administration of daily ketotifen started when the average tumor size reached 70 mm3 (day 18) and continued until completion of the study protocol. The first cycle of doxorubicin and anti–PD-L1 combination occurred when tumors reached an average size of 150 mm3 (day 22) and repeated on days 25, 28, and 31. The study ended when tumors reached a mean volume of 550 mm3 (day 33). Mice were sacrificed and tumors were collected for immunofluorescence staining experiments. Planar dimensions (x, y) of the tumor were monitored every 2 to 3 days using a digital caliper, and tumor volume was estimated from the volume of a sphere with a diameter equal to the average of planar dimensions.

Mice rechallenge

Mice were considered long-term survivors if no tumor was detected 80 days after the surgical resection of MCA205 primary tumors. Five long-term survivors from each treatment group were rechallenged with a subcutaneous injection of 2.5 × 105 MCA205 cells at the flank region. Naïve mice of the same age were injected in parallel to serve as controls.

Ethics approval

The clinical study of ketotifen (EudraCT #: 2022-002311-39) was approved in the European Union/European Economic Area under the Clinical Trials Directive 2001/20/EC and conducted in accordance with the Declaration of Helsinki regarding ethical principles for medical research involving human subjects. A written informed consent form was obtained from each participant included in the study and the study was approved by the Cyprus National Bioethics Committee (ΕΕΒΚ/ΕΠ /2022/36) and the Cyprus Pharmaceutical Services (ΚΔ 3/22).

Clinical ultrasound imaging

Ultrasound measurements of elastography from patients were collected as a component of an ongoing proof-of-concept phase II clinical trial (EudraCT #: 2022-002311-39), which aims to explore the impact of ketotifen on reducing tumor stiffness and improving the efficacy of chemotherapy in patients with sarcoma. Written informed consent was obtained from all participants.

Patients with different sarcoma subtypes were subjected to shear wave elastography (SWE) imaging before initiating ketotifen treatment to establish the baseline stiffness of the tumor. Each imaging session involved capturing five SWE images at different planes, and the average value of these images was considered. Subsequently, ultrasound imaging was conducted at regular intervals to monitor potential changes in stiffness induced by ketotifen administration.

Statistical analysis and reproducibility

Statistical analysis was performed using GraphPad Prism 9.0 software (RRID:SCR_002798). For tumor growth curves and ultrasound measurements involving multiple time points, two-way analyses of variants (ANOVA) followed by Dunnett multiple comparison were used. For other experiments including toxicity assessment, histology, and flow cytometry, one-way ANOVA or unpaired parametric Welch t test was used depending on the number of groups being compared. Only statistically significant differences along with the exact P values are displayed in the figures. A P value of ≤0.05 was considered statistically significant. Sample size and repetition of each experiment are indicated in corresponding figure captions. Sample size was based on previously published studies measuring the same output variables. Data are presented as means with standard errors.

Data availability

All other data supporting the findings of this study are available from the corresponding author and the clinicians responsible for running the clinical study upon request due to patient privacy. Additional methods used in the study can be found in Supplementary Data.

Mast cell degranulation promotes myofibroblast differentiation

The presence of MCs in sarcoma tumors was confirmed by CD117 (c-Kit) and tryptase immunofluorescence staining of human tissue microarrays. Tryptase is a serine protease unique to MCs with a reported role in myofibroblast proliferation and collagen synthesis (41, 44, 45). MCs were found in all sarcoma subtypes tested (Supplementary Fig. S1A) and they were at a closer distance to myofibroblasts in tumor tissue as compared with healthy tissue (Fig. 1A). To this end, we sought to investigate the consequences of MC activation on myofibroblasts by using a coculture assay whereby murine MC/9 MCs and NIH3T3 fibroblasts were separated by a transwell chamber with micropores that only allowed chemical communication (Fig. 1B). In this system, we determined MC degranulation efficiency by measuring the concentration of β-hexosaminidase (β-hex) released in the supernatant (46). MC degranulation was induced through exposure to compound 48/80 (C48/80), which binds to the MRGPRB2 receptor (the orthologue of the human G-protein–coupled receptor MRGPRX2; Supplementary Fig. S2A; ref. 47). Transforming growth factor β (TGFβ) was used as a fibroblast activator due to its capacity to induce the myofibroblast phenotype of NIH3T3 cells (48). MC/9 cells were cultivated with NIH3T3 cells for 24 hours and then treated with either ketotifen or TGFβ for an additional 24 hours in a low serum medium, followed by a 30-minute sensitization with C48/80. We found that ketotifen inhibits degranulation in a dose-dependent manner, with the concentration of 100 μg/mL causing more than 50% reduction in β-hex release compared with C48/80 treatment (Fig. 1C; Supplementary Fig. S2B and S2C). The presence of TGFβ in the coculture did not affect MC degranulation (Fig. 1C). To further explore the MC–fibroblast interaction, we monitored the differentiation process of NIH3T3 cells in the coculture system by assessing the levels of αSMA and Collagen I, which are two markers of the myofibroblast phenotype (Fig. 1DF). A significant increase in both αSMA and Collagen I protein expression occurred only in the presence of both TGFβ and activated MCs, whereas no change was observed in the presence of TGFβ alone. On the contrary, ketotifen impaired this myofibroblast differentiation (Fig. 1DF). The effect of MCs on myofibroblast activity was further confirmed in CAFs isolated from murine fibrosarcoma tumors (MCA205) and cocultured with MC/9 cells (Supplementary Fig. S1B). RNA isolation from CAFs and assessment of fibrosis-related genes, such as Col3A1 and CTGF, indicated a significant upregulation in transcript levels upon exposure to MCs, whereas the addition of ketotifen inverted this effect. MCs, however, did not have any impact on regulating the expression of αSMA (ACTA2) and collagen I (Col1A1; Supplementary Fig. S1C–S1F). Therefore, our findings demonstrate that ketotifen can directly affect CAF function by antagonizing H1 receptor binding and indirectly by stabilizing MCs and impeding their degranulation.

Figure 1.

Inhibition of mast cell degranulation suppresses myofibroblast differentiation and activity. A, Representative fluorescence images of human tissue arrays showing the proximity of tryptase-positive mast cells (green color) to αSMA-positive CAFs (red color) in tumor tissue (top) as compared with normal tissue (bottom); scale bar = 0.2 mm (n = 4–6 human tissue arrays). B, Transwell coculture system used for the in vitro evaluation of MC/9 mast cells and NIH3T3 fibroblasts interaction. C, Inhibition of MC/9 degranulation after exposure to ketotifen. For the analysis of MC degranulation, we measured the release of β-hexosaminidase into the culture medium (n = 3 independent experiments, N = 6 technical replicates per experiment). D, Images of immunofluorescence staining of αSMA and collagen I in fibroblasts; scale bar = 0.1 mm. E, Quantification of αSMA and (F) collagen I–positive staining normalized to DAPI nuclear staining. Data, mean ± SE. For C, E, and F, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test. (B, Created with BioRender.com.)

Figure 1.

Inhibition of mast cell degranulation suppresses myofibroblast differentiation and activity. A, Representative fluorescence images of human tissue arrays showing the proximity of tryptase-positive mast cells (green color) to αSMA-positive CAFs (red color) in tumor tissue (top) as compared with normal tissue (bottom); scale bar = 0.2 mm (n = 4–6 human tissue arrays). B, Transwell coculture system used for the in vitro evaluation of MC/9 mast cells and NIH3T3 fibroblasts interaction. C, Inhibition of MC/9 degranulation after exposure to ketotifen. For the analysis of MC degranulation, we measured the release of β-hexosaminidase into the culture medium (n = 3 independent experiments, N = 6 technical replicates per experiment). D, Images of immunofluorescence staining of αSMA and collagen I in fibroblasts; scale bar = 0.1 mm. E, Quantification of αSMA and (F) collagen I–positive staining normalized to DAPI nuclear staining. Data, mean ± SE. For C, E, and F, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test. (B, Created with BioRender.com.)

Close modal

Ketotifen inhibits MCs in sarcoma and reduces tumor stiffness

Following the confirmation of the presence of MCs in human sarcoma samples and the in vitro characterization of MC–fibroblast interaction, we hypothesized that inhibition of MC degranulation would normalize ECM in murine sarcoma models. We assessed the impact of MC degranulation by treating MCA205 fibrosarcoma and K7M2 osteosarcoma tumors with ketotifen. Ketotifen was administered in four different doses (i.e., 1, 5, 10, and 25 mg/kg), once a day by intraperitoneal injection for a period of 8 days (refs. 49–51; Fig. 2A). Although no dose exhibited an antitumor effect (Supplementary Fig. S3A–S3D; ref. 52), the highest dose caused splenomegaly (Supplementary Fig. S4). To confirm the effect of ketotifen on MC degranulation, we stained ketotifen-treated tumors for tryptase (41, 44, 45) and found reduced levels after treatment with 10 mg/kg ketotifen in both sarcoma models (Supplementary Fig. S3E–S3G).

Figure 2.

Ketotifen alleviates tumor stiffness. A, Study treatment protocol for the MCA205 and K7M2 tumor models. For the MCA205 model, ketotifen was administered at 1, 5, 10, and 25 mg/kg daily via intraperitoneal injection for 8 days once tumor size reached an average volume of 60 mm3. For the K7M2 model, ketotifen was administered at 10 mg/kg daily via intraperitoneal injection for 8 days once tumor size reached an average volume of 150 mm3. B, Representative SWE images of MCA205 tumors treated with saline and ketotifen 10 mg/kg. The dashed line denotes the tumor region, and the color map indicates the different magnitudes of elastic modulus in kPa. C, Temporal changes in elastic modulus of MC205 and (D) K7M2 tumors during the treatment protocol. Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two-way ANOVA test (n = 5 mice, N = 2 image fields per mouse, each point represents the average value of 2 images). E, Table with demographics and the effect of ketotifen on tumor stiffness in sarcoma patients. F, Representative SWE images of tumors pre- and post-ketotifen treatment. G, Graph showing the tumor elastic modulus before and after ketotifen treatment for each patient. Data, mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test. (A, Created with BioRender.com.)

Figure 2.

Ketotifen alleviates tumor stiffness. A, Study treatment protocol for the MCA205 and K7M2 tumor models. For the MCA205 model, ketotifen was administered at 1, 5, 10, and 25 mg/kg daily via intraperitoneal injection for 8 days once tumor size reached an average volume of 60 mm3. For the K7M2 model, ketotifen was administered at 10 mg/kg daily via intraperitoneal injection for 8 days once tumor size reached an average volume of 150 mm3. B, Representative SWE images of MCA205 tumors treated with saline and ketotifen 10 mg/kg. The dashed line denotes the tumor region, and the color map indicates the different magnitudes of elastic modulus in kPa. C, Temporal changes in elastic modulus of MC205 and (D) K7M2 tumors during the treatment protocol. Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two-way ANOVA test (n = 5 mice, N = 2 image fields per mouse, each point represents the average value of 2 images). E, Table with demographics and the effect of ketotifen on tumor stiffness in sarcoma patients. F, Representative SWE images of tumors pre- and post-ketotifen treatment. G, Graph showing the tumor elastic modulus before and after ketotifen treatment for each patient. Data, mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test. (A, Created with BioRender.com.)

Close modal

We also monitored the effect of ketotifen on tumor stiffness in vivo using ultrasound SWE (Fig. 2B). We performed SWE measurements before, after 3 days (for MCA205) or 4 days (for K7M2) of ketotifen treatment, and upon study completion. Our results demonstrate that the 10 mg/kg dose reduced tissue stiffness most effectively in MCA205 fibrosarcoma tumors, with elastic modulus values reaching 20 kPa (Fig. 2C). K7M2 osteosarcoma tumors similarly benefited from 10 mg/kg of daily ketotifen (Fig. 2D). The pertinent reduction in tumor stiffness was further validated ex vivo at the microscopic tissue scale using atomic force microscopy (AFM) of tumor samples. AFM analysis indicated a reduction in the average value of elastic modulus of K7M2 tumors (Supplementary Fig. S3H), with a shifted distribution toward lower values, which suggests a decrease in collagen density (Supplementary Fig. S3I; ref. 53). Notably, the effect of ketotifen is transient as 14 days following the last day of ketotifen administration, the tumor acquires the stiffness level of the control group (Supplementary Fig. S3J).

Next, we sought to assess the clinical value of ketotifen in reprogramming the TME and reducing stiffness. To achieve this, we analyzed preliminary data from the ongoing phase II clinical trial investigating the use of ketotifen to augment therapy in sarcoma patients. A total of 8 patients with various sarcoma subtypes, including epithelioid leiomyosarcoma, myxofibrosarcoma, atypical lipomatous tumor, leiomyosarcoma, pleomorphic leiomyosarcoma, synovial sarcoma, and desmoid tumor, underwent initial ultrasound imaging before ketotifen treatment to establish the baseline tissue stiffness. Subsequently, elastography measurements were taken throughout the course of ketotifen administration for patients who either received systemic therapy or patients who underwent surgery alone (Fig. 2E).

Ketotifen was administered at the recommended dose of 1 mg twice daily to all patients. Notably, in all experimental cases, tumor stiffness significantly decreased upon ketotifen treatment, even though the baseline tissue stiffness was notably lower compared with the mouse studies (Fig. 2FG).

MCs deactivate CAFs inducing ECM remodeling

As tissue stiffening is a consequence of collagen abundance in the intratumoral space (54), and based on our in vitro study on MC–fibroblast cross-talk, we hypothesized that the activity of CAFs will also be affected by ketotifen intervention, either directly or indirectly. We found that only the concentration of 10 mg/kg ketotifen reduced both CAF density and proliferation, as measured by the expression of αSMA and Ki67, respectively (Fig. 3A and B; Supplementary Fig. S5A and S5B). We observed that the administration of the highest dose of ketotifen did not affect the proliferation of CAFs. This suggests that the tumor may trigger secondary responses in an effort to compensate for and maintain the fibrotic stroma. The reduction of αSMA was also confirmed on transcript level along with a decrease in collagen mRNA and protein levels (Fig. 3CF). Besides collagen, CAFs synthesize hyaluronic acid, suggesting that changes in CAF activation and proliferation could result in inhibition of hyaluronan production. To address this, we performed immunofluorescence staining for hyaluronan binding protein 1 (HABP1), which demonstrated a suppression in hyaluronan levels at the optimum dose of ketotifen (Fig. 3G and H; Supplementary Fig. S5C and S5D).

Figure 3.

Ketotifen reprograms the TME by normalizing CAFs, collagen, and hyaluronan. A, Representative immunofluorescence images of Ki67 proliferation marker (red) and αSMA (green) in MCA205 fibrosarcoma tumors; scale bar = 0.2 μm. B, Quantification of CAFs positive for both the αSMA and Ki67 markers, normalized to total αSMA staining. C, Quantification of mRNA expression levels of Col1A1 and ACTA2 (i.e., the gene encoding αSMA) in untreated and ketotifen (10 mg/kg) treated MCA205 tumors using the 2−ΔΔCT method (3 biological × 3 technical replicates were used). D, Representative bright field images of picrosirius red staining in MCA205 paraffin sections; scale bar, 0.5 mm. E, Quantification of the area positive for picrosirius red staining in MCA205 fibrosarcoma and (F) K7M2 osteosarcoma tumors. G, Representative immunofluorescence images of MCA205 paraffin sections stained with anti-HABP1; scale bar, 0.2 mm. H, Quantification of the fraction of area positive for HABP1 staining (green) normalized to DAPI (blue) stain in MCA205 tumors. For histologic analysis, n = 4 tumors per group were stained, and N = 5–6 image fields were captured per tumor. Data are presented as mean ± SE. For B, C, E, and H, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test and for F using the unpaired parametric Welch t test.

Figure 3.

Ketotifen reprograms the TME by normalizing CAFs, collagen, and hyaluronan. A, Representative immunofluorescence images of Ki67 proliferation marker (red) and αSMA (green) in MCA205 fibrosarcoma tumors; scale bar = 0.2 μm. B, Quantification of CAFs positive for both the αSMA and Ki67 markers, normalized to total αSMA staining. C, Quantification of mRNA expression levels of Col1A1 and ACTA2 (i.e., the gene encoding αSMA) in untreated and ketotifen (10 mg/kg) treated MCA205 tumors using the 2−ΔΔCT method (3 biological × 3 technical replicates were used). D, Representative bright field images of picrosirius red staining in MCA205 paraffin sections; scale bar, 0.5 mm. E, Quantification of the area positive for picrosirius red staining in MCA205 fibrosarcoma and (F) K7M2 osteosarcoma tumors. G, Representative immunofluorescence images of MCA205 paraffin sections stained with anti-HABP1; scale bar, 0.2 mm. H, Quantification of the fraction of area positive for HABP1 staining (green) normalized to DAPI (blue) stain in MCA205 tumors. For histologic analysis, n = 4 tumors per group were stained, and N = 5–6 image fields were captured per tumor. Data are presented as mean ± SE. For B, C, E, and H, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test and for F using the unpaired parametric Welch t test.

Close modal

Improved perfusion and oxygenation by restoration of vessel functionality

Increased tumor stiffness is indicative of the accumulation of intratumoral mechanical forces that compress blood vessels and, thus, induce hypoperfusion (55–57). There is also evidence that MCs are angiogenic mediators, as they increase vascular permeability (58, 59) and neovascularization (60, 61), which could also reduce blood flow. Accordingly, we hypothesized that MC inhibition could restore the function of tumor blood vessels by both reducing vessel permeability and decompressing the vessels. Indeed, administration of ketotifen at 5, 10, and 25 mg/kg increased the pericyte coverage of the tumor vasculature that fortifies the vessel walls, as indicated by the colocalization of the NG2 pericyte marker and CD31 endothelial protein, compared with the lowest dose and untreated control. Notably, the vascular density was not affected (Fig. 4AC). Surprisingly, we did not observe any reduction in VEGF transcripts. Instead, we observed an upregulation in IFNγ transcription levels in MCA205 tumors following 10 mg/kg ketotifen treatment (Fig. 4D). IFNγ is an angiostatic protein whose concentration in the TME has been directly related to vessel normalization and reduced vessel leakiness (62–64). Furthermore, the reduction in IFP levels with ketotifen treatment provides another evidence of vascular normalization (Fig. 4E). In line with this, ketotifen increased the fraction of vessels with open lumen, a proof of vessel decompression (Fig. 4F).

Figure 4.

Ketotifen restores tumor vessel functionality and perfusion. A, Representative immunofluorescence images of NG2 pericyte marker (green) and CD31 endothelial cell marker (red) of MCA205 tumors upon treatment with different ketotifen concentrations; scale bar = 0.1 mm. B, Quantification of vessel pericyte coverage as indicated by CD31 and NG2 overlapping staining (yellow) normalized to total CD31+ staining. C, Quantification of area fraction positive for CD31 staining (vessels; n = 5 mice per group, N = 3–5 image fields per mouse). D, Quantification of mRNA expression levels of IFNγ and VEGF in untreated and ketotifen (10 mg/kg) treated MCA205 tumors using the 2−ΔΔCT method (3 biological × 3 technical replicates were used). E, Interstitial fluid pressure levels in untreated and daily ketotifen-treated mice for 7 days (n = 7 mice per treatment group). F, Quantification of open lumen fraction in MCA205 tumors based on CD31 image analysis. G, Representative contrast-enhanced ultrasound images of microbubbles (yellow) entering MCA205 tumors for control and 10 mg/kg ketotifen-treated tumors at the time of peak intensity. The dashed line shows the tumor margin. H, Normalized perfused area with respect to the total tumor area at the time of peak intensity for MCA205 and (I) K7M2 tumors, after completion of treatment protocol (Fig. 2A; n = 4–5). J, Representative immunofluorescence images of MCA205 paraffin sections stained for pimonidazole adducts following pimonidazole hydrochloride injection; scale bar = 0.2 mm. Quantification of hypoxia area fraction (red) normalized to DAPI stain (blue) of (K) MCA205 and (L) K7M2 tumors (n = 5 mice per group, N = 3–5 image fields per mouse). Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two independent groups using the ordinary one-way ANOVA test for B, DF, H, KL and the unpaired parametric Welch t test for C and I.

Figure 4.

Ketotifen restores tumor vessel functionality and perfusion. A, Representative immunofluorescence images of NG2 pericyte marker (green) and CD31 endothelial cell marker (red) of MCA205 tumors upon treatment with different ketotifen concentrations; scale bar = 0.1 mm. B, Quantification of vessel pericyte coverage as indicated by CD31 and NG2 overlapping staining (yellow) normalized to total CD31+ staining. C, Quantification of area fraction positive for CD31 staining (vessels; n = 5 mice per group, N = 3–5 image fields per mouse). D, Quantification of mRNA expression levels of IFNγ and VEGF in untreated and ketotifen (10 mg/kg) treated MCA205 tumors using the 2−ΔΔCT method (3 biological × 3 technical replicates were used). E, Interstitial fluid pressure levels in untreated and daily ketotifen-treated mice for 7 days (n = 7 mice per treatment group). F, Quantification of open lumen fraction in MCA205 tumors based on CD31 image analysis. G, Representative contrast-enhanced ultrasound images of microbubbles (yellow) entering MCA205 tumors for control and 10 mg/kg ketotifen-treated tumors at the time of peak intensity. The dashed line shows the tumor margin. H, Normalized perfused area with respect to the total tumor area at the time of peak intensity for MCA205 and (I) K7M2 tumors, after completion of treatment protocol (Fig. 2A; n = 4–5). J, Representative immunofluorescence images of MCA205 paraffin sections stained for pimonidazole adducts following pimonidazole hydrochloride injection; scale bar = 0.2 mm. Quantification of hypoxia area fraction (red) normalized to DAPI stain (blue) of (K) MCA205 and (L) K7M2 tumors (n = 5 mice per group, N = 3–5 image fields per mouse). Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two independent groups using the ordinary one-way ANOVA test for B, DF, H, KL and the unpaired parametric Welch t test for C and I.

Close modal

Next, we sought to investigate whether restoration of these tumor blood vessel abnormalities was sufficient to reestablish tumor blood perfusion and improve oxygenation. Contrast-enhanced ultrasound imaging shows an increase in the area of the tumor covered by the contrast agent (Fig. 4G). Our findings indicate that 10 mg/kg ketotifen was the most effective dose at increasing vessel perfusion in both fibrosarcoma and osteosarcoma tumors (Fig. 4H and I). Similarly, by measuring the intratumoral levels of hypoxia using pimonidazole, we observed that 10 mg/kg ketotifen alleviated hypoxia in both tumor models (Fig. 4JL). We conclude that ketotifen at a daily dose of 10 mg/kg can effectively reprogram MCs and CAFs, normalizing both the tumor ECM and vasculature and thus improving perfusion and oxygenation.

Improved effectiveness of chemoimmunotherapy and association with tumor stiffness

Subsequently, we set out to investigate the ability of ketotifen to improve chemoimmunotherapy in the same sarcoma models. To mimic the clinical therapeutic approach for patients with sarcoma, we used doxorubicin chemotherapy and the immune-checkpoint inhibitor, anti–PD-L1 antibody, after confirming its expression in tumor tissue (Supplementary Fig. S6A and S6B). First, we pretreated mice bearing MCA205 tumors with daily 10 mg/kg ketotifen for 3 days to restore vessel functionality and thus assist the delivery of anticancer treatment. The anticancer treatment administered was three cycles of 5 mg/kg doxorubicin and/or 10 mg/kg anti–PD-L1, every three days. In the K7M2 study, mice were administered ketotifen for four days, followed by four cycles of chemoimmunotherapy (Fig. 5A). Neither anti–PD-L1 nor doxorubicin monotherapy exhibited an antitumor activity as indicated by the growth curves of MCA205 and K7M2 tumors, whereas combination with ketotifen induced a significant antitumor response (Fig. 5B and C). The greatest decrease in tumor volume was observed when ketotifen was coadministered with the combination of doxorubicin and anti–PD-L1 antibody (Fig. 5B and C).

Figure 5.

Ketotifen enhances long-term response to chemoimmunotherapy combination treatment. A, Study treatment protocol for the MCA205 and K7M2 tumor model. Ketotifen was given daily until the completion of the study, once the tumors were palpable. For the MCA205 tumor model, the first cycle of doxorubicin or/and anti–PD-L1 (aPD-L1) treatment was administered via intraperitoneal injection on day 7, allowing ketotifen to prime the TME. Two additional cycles of doxorubicin or/and anti–PD-L1 were followed on days 10 and 13 for MCA205. Shear wave elastography measurements were obtained on day 7 right before chemoimmunotherapy initiation, on day 11, and after completion of the three treatment cycles (day 15). Primary tumors were excised on day 16. On day 63, survivors were rechallenged with the MCA205 cell line, and tumor volume was monitored for 24 days. For the K7M2 tumor model, doxorubicin or/and anti–PD-L1 treatment was administered on days 22, 25, 28, and 31. Shear wave elastography measurements were obtained on day 22 right before chemoimmunotherapy initiation and on day 32. Mice were sacrificed and primary tumors were excised on day 33. B, Growth curves of MCA205 tumors treated as indicated until day 16 (n = 10 mice per group). C, Relative growth curves of K7M2 tumors treated as indicated (n = 6 mice per group). Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two-way ANOVA test with Dunnett correction. D, Clustering of the experimental average tumor growth after treatment and elastic modulus. Error bars present the mean ± SD of clusters. E, Linear regression of tumor growth rate after treatment (Vr) as a function of elastic modulus (E), initial volume at the beginning of treatment (V0), tumor type, and treatment. Vr = a E + b V0 + c E V0 + d, where the parameters a, b, and c depend on tumor type and d on treatment and tumor type. The fitted model has a coefficient of regression (R2) equal to 0.79. F,Ex vivo immunofluorescence images of ketotifen or control K7M2 tumors 6 hours after the intravenous injection of the Atto 680-anti–PD-L1 antibody. G, Individual growth curves of MCA205 tumors after rechallenging survivors. Age-matched naïve mice were used as control. (A, Created with BioRender.com.)

Figure 5.

Ketotifen enhances long-term response to chemoimmunotherapy combination treatment. A, Study treatment protocol for the MCA205 and K7M2 tumor model. Ketotifen was given daily until the completion of the study, once the tumors were palpable. For the MCA205 tumor model, the first cycle of doxorubicin or/and anti–PD-L1 (aPD-L1) treatment was administered via intraperitoneal injection on day 7, allowing ketotifen to prime the TME. Two additional cycles of doxorubicin or/and anti–PD-L1 were followed on days 10 and 13 for MCA205. Shear wave elastography measurements were obtained on day 7 right before chemoimmunotherapy initiation, on day 11, and after completion of the three treatment cycles (day 15). Primary tumors were excised on day 16. On day 63, survivors were rechallenged with the MCA205 cell line, and tumor volume was monitored for 24 days. For the K7M2 tumor model, doxorubicin or/and anti–PD-L1 treatment was administered on days 22, 25, 28, and 31. Shear wave elastography measurements were obtained on day 22 right before chemoimmunotherapy initiation and on day 32. Mice were sacrificed and primary tumors were excised on day 33. B, Growth curves of MCA205 tumors treated as indicated until day 16 (n = 10 mice per group). C, Relative growth curves of K7M2 tumors treated as indicated (n = 6 mice per group). Data are presented as mean ± SE. Statistical analyses were performed by comparing means between two independent groups using the two-way ANOVA test with Dunnett correction. D, Clustering of the experimental average tumor growth after treatment and elastic modulus. Error bars present the mean ± SD of clusters. E, Linear regression of tumor growth rate after treatment (Vr) as a function of elastic modulus (E), initial volume at the beginning of treatment (V0), tumor type, and treatment. Vr = a E + b V0 + c E V0 + d, where the parameters a, b, and c depend on tumor type and d on treatment and tumor type. The fitted model has a coefficient of regression (R2) equal to 0.79. F,Ex vivo immunofluorescence images of ketotifen or control K7M2 tumors 6 hours after the intravenous injection of the Atto 680-anti–PD-L1 antibody. G, Individual growth curves of MCA205 tumors after rechallenging survivors. Age-matched naïve mice were used as control. (A, Created with BioRender.com.)

Close modal

To support our hypothesis that the increased efficacy of cytotoxic treatment was attributed to ketotifen effects, we compared the average tumor elastic modulus measured with SWE with the average tumor growth rate from the first cycle of chemoimmunotherapy until the end of the experiment in both MCA205 and K7M2 tumor models, and we examined their clustering pattern (65). We used an unsupervised learning methodology to process the data, which separated them into four clusters: a cluster with low elastic modulus and low tumor growth rate (cluster 1), a cluster with low elastic modulus and high growth rate (cluster 2), a cluster with high elastic modulus and low tumor growth rate (cluster 3), and a cluster with high elastic modulus and high tumor growth rate (cluster 4; Fig. 5D; Supplementary Fig. S6C). Clusters 1 and 2 consist solely of mice treated with ketotifen either with or without anticancer therapy. Specifically, ketotifen monotherapy group corresponds to 4% of total mice in cluster 1, whereas in cluster 2, ketotifen monotherapy accounts for 69%, suggesting that ketotifen alone is less likely to cause an antitumor effect, but the combination of ketotifen, doxorubicin, and anti–PD-L1 belongs solely to cluster 1 with a low tumor growth rate. Interestingly, there are cases in cluster 1 that have a negative tumor growth rate. Half of mice comprising cluster 3 received anti–PD-L1 plus doxorubicin combination, whereas most mice in cluster 4 correspond to the control group followed by anti–PD-L1, doxorubicin monotherapies or combination.

Furthermore, a linear regression analysis was performed to generate a model capable of predicting the average tumor growth after a specific treatment or combination of treatments (Fig. 5E; Supplementary Tables S1 and S2). The model is a function of the elastic modulus (E), initial volume at the beginning of treatment ( |${{V}_0}$|⁠), tumor type and treatment ( |${{V}_r}\ = \ a\ E + b\ {{V}_0} + c\ E\ {{V}_0} + d$|⁠), where the parameters a, b|$,$| and c depend on tumor type and d on treatment and tumor type. Parameter d (mm3/day) directly affects tumor growth rate and can be used as a means of comparison for every group that has similar E and |${{V}_0}$|⁠. For instance, parameter d of doxorubicin monotherapy in MCA205 and K7M2 tumor models is reduced by 10% and 21.1%, respectively, compared with the control groups ( |$d\ = $| −162.6 for MCA205, |$d\ = \ 57.7$| for K7M2). Upon comparison of control with anti–PD-L1 monotherapy with regard to parameter d, the changes are minor, with a 2% increase and 7.5% decrease for the MCA205 and K7M2 tumor models, respectively. On the contrary, by comparing parameter d of the ketotifen group ( |$d\ = $| −35.1 for MCA205, |$d\ = $| 62.7 for K7M2) with that of the ketotifen + doxorubicin + anti–PD-L1 group, we observed a 138.4% and 74.36% reduction for MCA205 and K7M2 tumor models, respectively. According to the ANOVA test, the coefficient dd of anti–PD-L1 and doxorubicin are significantly different than zero, whereas the coefficient d of ketotifen alone does not differ from zero. However, the coefficient d of ketotifen + doxorubicin is significantly different than zero, which indicates additive effects. The capability of ketotifen to remodel the tumor stroma and subsequently to promote the efficacy of drugs, such as ICIs, is also evident when measuring the intratumoral delivery of anti–PD-L1 antibodies conjugated with the fluorescence probe Atto 680 (Fig. 5F). K7M2 tumor-bearing mice pretreated with ketotifen exhibit a higher accumulation of anti–PD-L1 in the tumor rather than the control mice.

Subsequently, we investigated if this immune response is preserved against rechallenge of long-term survivors with the same cell line. Mice were considered long-term survivors if no tumor was detected 47 days after the surgical resection of MCA205 primary tumors. Five long-term survivors from each treatment group were rechallenged with a subcutaneous injection of 2.5 × 105 MCA205 cells at the flank region, whereas naïve mice of the same age were injected in parallel to serve as controls. Interestingly, no tumor growth occurred in the ketotifen + doxorubicin + anti–PD-L1 group, suggesting the development of an adaptive memory response, which is triggered upon encountering tumor antigens. On the contrary, all naïve and ketotifen-treated mice developed progressively growing tumors. Likewise, large tumors were formed in three mice of the doxorubicin + anti–PD-L1 group and in four mice of the anti–PD-L1 group. In the ketotifen + doxorubicin and ketotifen + anti–PD-L1 group, only one mouse out of five developed fibrosarcoma (Fig. 5G). In agreement with the hierarchy of efficacy observed in tumor growth measurements, the combination of ketotifen + doxorubicin + anti–PD-L1 demonstrated increased immunologic memory compared with the doxorubicin + anti–PD-L1 group.

Taken together, these findings demonstrate that pretreatment with ketotifen can effectively establish favorable conditions within the TME toward enabling doxorubicin and anti–PD-L1 antibody treatment to confer immunologic memory.

Effects of ketotifen on reprogramming the immune TME

To further evaluate the contribution of ketotifen in stimulating immune responses, we assessed the presence of T-cell populations in MCA205 tumors using flow cytometry. Total T-cell population was primarily defined by the expression of CD3+ protein, a pan T-cell marker, and then distinguished into the cytotoxic CD8+ or the regulatory T-cell subtype (CD3+CD4+CD25hiCD127loFoxp3+; Fig. 6A; Supplementary Fig. S8A). We observed a statistically significant increase in total T-cell recruitment of the doxorubicin + anti–PD-L1 and ketotifen + doxorubicin + anti–PD-L1 treatment groups. In K7M2 tumors, however, only the groups of mice receiving ketotifen prior to cytotoxic treatment exhibited an increase in T-cell infiltration as indicated by immunofluorescence staining for CD3 protein (Supplementary Fig. S7D). Moreover, we found that the ratio of CD8+ T cells to immunosuppressive Tregs was significantly increased after combining ketotifen with doxorubicin and immunotherapy (Fig. 6B). Immunofluorescence staining for Ki67 and CD8+ confirmed the activation status of recruited CD8+ T cells in osteosarcoma tumors (Fig. 6C and D). Considering that ketotifen upregulates IFNγ, which is a key mediator of antitumor immunity with vascular normalization activity (66–69), we hypothesized that accumulation of CD8+ T cells achieved by ketotifen combination with either doxorubicin or PD-L1 blockade is the result of tissue reoxygenation. To address this, we stained K7M2 tissue sections for pimonidazole adducts as a measure of hypoxia formation and found that only the groups receiving ketotifen exhibited a significant reduction of hypoxic areas allowing for adequate tumor oxygenation (Supplementary Fig. S7A and S7B). We further explored if restoration of intratumoral T-cell infiltration occurs via an upregulation of immune cell adhesion to the wall of perfused vessels. Ketotifen treatment increased mRNA expression of the immune cell adhesion molecules ICAM and P-selectin (P-sel) (Supplementary Fig. S7C). We confirmed these findings by immunofluorescence staining of CD3 and CD31 endothelial markers demonstrating that all combination treatments of ketotifen exhibited more association between T lymphocytes and endothelial cells, without affecting the endothelial cell area fraction (Supplementary Fig. S7E). Consistent with previous studies highlighting the effect of ICI on improving vascular perfusion (70), we found that anti–PD-L1 monotherapy and doxorubicin combination increase the colocalization of CD3+ T cells and CD31 (Fig. 6E and F). Furthermore, as hypoxia fosters the accumulation of myeloid stroma and polarization into suppressive cell populations, such as myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages, we confirmed that disruption of hypoxic zones by ketotifen has an immediate effect on potentiating the antitumor immune responses of doxorubicin and immunotherapy by reducing MDSC density in MCA205 tumors (Supplementary Fig. S8B and S8C). Unlike other studies supporting that doxorubicin monotherapy decreases the number of MDSCs, leading to enhanced numbers of CD4+ and CD8+ cells (71, 72), here, we did not observe such a reduction and the immunologic benefit of the doxorubicin + ICI combination was relatively modest and did not counterweight that induced upon ketotifen combination.

Figure 6.

Ketotifen pretreatment enhances the antitumor effects of immunotherapy by promoting T-cell recruitment and cytotoxic immune responses. A, Flow cytometry data. Percentage of the total CD3+ T cells among CD45+ lymphocytes in the whole tumor tissue of MCA205 tumor models treated as indicated. B, Flow cytometry data. Ratio of cytotoxic CD3+CD8+ T cells to CD3+CD4+CD25hiCD127loFoxp3+ Tregs (n = 5 mice per group). C, Representative images of IF staining of CD8 (green) and Ki67 (red) in K7M2 paraffin-embedded tissue sections; scale bar = 0.1 mm. D, Quantification of proliferative CD8+ T-cell fraction as the ratio of area that is double positive for CD8 and Ki67 staining (yellow) to the area that is positive for total CD8 staining (n = 4 mice per group, N = 4–5 image fields per mouse). E, Representative images of IF staining of CD31 (endothelial marker, red) and CD3 (green) in K7M2 frozen tissue sections; scale bar = 0.1 mm. F, Quantification of colocalization CD3+ T cells with CD31+ endothelial cells normalized to the total CD31-positive staining (n = 3 mice per group, N = 3–5 image fields per mouse). Data are presented as mean ± SE. For A, B, D, and F, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test.

Figure 6.

Ketotifen pretreatment enhances the antitumor effects of immunotherapy by promoting T-cell recruitment and cytotoxic immune responses. A, Flow cytometry data. Percentage of the total CD3+ T cells among CD45+ lymphocytes in the whole tumor tissue of MCA205 tumor models treated as indicated. B, Flow cytometry data. Ratio of cytotoxic CD3+CD8+ T cells to CD3+CD4+CD25hiCD127loFoxp3+ Tregs (n = 5 mice per group). C, Representative images of IF staining of CD8 (green) and Ki67 (red) in K7M2 paraffin-embedded tissue sections; scale bar = 0.1 mm. D, Quantification of proliferative CD8+ T-cell fraction as the ratio of area that is double positive for CD8 and Ki67 staining (yellow) to the area that is positive for total CD8 staining (n = 4 mice per group, N = 4–5 image fields per mouse). E, Representative images of IF staining of CD31 (endothelial marker, red) and CD3 (green) in K7M2 frozen tissue sections; scale bar = 0.1 mm. F, Quantification of colocalization CD3+ T cells with CD31+ endothelial cells normalized to the total CD31-positive staining (n = 3 mice per group, N = 3–5 image fields per mouse). Data are presented as mean ± SE. For A, B, D, and F, statistical analyses were performed by comparing means between two independent groups using the ordinary one-way ANOVA test.

Close modal

Sarcomas comprise a heterogenous group of malignancies, of more than 100 different subtypes, arising from mesenchymal tissue. Despite the recent advances in cancer research, the application of immunotherapy in daily clinical practice for the treatment of sarcoma remains limited. Clinical studies have yielded conflicting results demonstrating that blockade of the PD-1/PD-L1 axis can benefit only specific histologic subtypes (4), thus highlighting the need for alternative treatment strategies. Given the role of the TME in determining the behavior and response to treatment, the current study exploited the potential of stabilizing the intratumoral MCs using the common antihistamine drug ketotifen. We primarily demonstrated that ketotifen is capable of inhibiting MC degranulation by stabilizing their membranes and, thus, making them less likely to rupture and release their content. We showed that blocking of granular content can subsequently impact fibroblast phenotype by reducing the expression of αSMA and collagen synthesis as indicated in the coculture experiments. Although the exact mechanisms are not fully elucidated, MCs can influence fibroblast differentiation through the release of various mediators and direct cell-to-cell interactions within the TME. Furthermore, we showed that ketotifen can exert a direct effect on CAFs by antagonizing H1 receptor binding on the cell surface and causing the subsequent inhibition of intracellular signaling events leading to the downregulation of ECM-related genes. In addition to histamine, MCs might release additional mediators, including TGFβ, IL13, and tryptase, which contribute to tumor fibrosis (73). TGFβ can promote fibroblast migration, proliferation, collagen formation, and fibroblast differentiation into CAF. IL13 is also a profibrotic mediator, which can promote fibrosis via TGFβ-dependent and independent mechanisms. Tryptase stimulates fibroblast proliferation and synthesis of collagen and fibronectin via the activation of the protease-activated receptor PAR-2 (74). Although not yet confirmed in the context of tumors, CCL2 also produced by MCs has been reported to attract fibrocytes to regions of injury (75).

Furthermore, we note that in this study, we focused on myofibroblastic CAFs, which are the major contributors to fibrosis, whereas a more thorough analysis comparing the single-cell transcriptome of the untreated and the ketotifen-treated groups is required to appreciate the full spectrum of fibroblast heterogeneity in solid tumors and to develop therapies that specifically target tumor-promoting CAFs. In line with this, a recent analysis of the TME of pancreatic tumors has demonstrated a distinct CAF subpopulation (CD141+MHCII) as the responsible cell population of desmoplasia and a potential therapeutic target. Indeed, the CD141+MHCII CAF population resembled the myofibroblast subtype with regard to gene expression and active collagen production but also shared markers with other subtypes including the inflammatory CAFs (76). Another study suggests that for invasive squamous tumors of the pancreas exhibiting a higher proportion of activated stromal cells, blockade of the activated stroma using FAP inhibitors should be considered in the treatment regimen for augmented therapeutic outcome (77). Thus, a therapeutic strategy can be designed based on the characteristics of CAF subtypes.

The MC–CAF cellular cross-talk was further validated in murine sarcoma models. By titrating the ketotifen dose, we found that the dose of 10 mg/kg had the most prominent effect on tissue stiffness via reducing CAF proliferation and density. Consistent with the in vitro results, collagen and hyaluronan synthesis was also suppressed, suggesting that ketotifen exerts its TME reprogramming properties by affecting CAFs. Importantly, the resulting alleviation of stiffness enhanced vessel decompression, allowing for improved perfusion and oxygen delivery to the tumor site. Interestingly, ketotifen was also able to fortify the tumor vessel walls by increasing pericyte coverage.

Approved drugs that can reprogram/normalize the TME have already been repurposed and entered clinical trials, holding great promise in broadening the accessibility and increasing the efficacy of immunotherapies (78–81). Recent findings indicated that patients of multiple tumor types, who took H1-antihistamines during immune-checkpoint blockade treatment had a highly significantly reduced death rate compared with those who did not, whereas antihistamines had a minimal effect on the survival of chemotherapy-treated patients, which further supports the immunologic effects of ketotifen rather than any direct antitumor effects (82, 83). Accordingly, in our attempt to improve the responsiveness of fibrosarcoma and osteosarcoma tumors to conventional therapies, we combined the reprogramming properties of ketotifen with the cytotoxic activities of doxorubicin and anti–PD-L1. We found that pretreatment with ketotifen increases the sensitivity of MCA205 and K7M2 tumors to chemotherapy and checkpoint inhibition, whereas the combination of the three augments antitumor response as indicated by the regression of tumor volume. Doxorubicin acts by intercalating within the DNA and directly killing cancer cells, whereas antibodies that block the signal from T-cell coinhibitory receptors exert their antitumor responses by resuscitating the host immune system and, thus, leading to a more durable response (84–86). Another possible mechanism by which ketotifen may sensitize cancer cells to doxorubicin is via inhibition of exosome release. Although not explored in this study, ketotifen administration can inhibit calcium entry into the cell and in turn reduce exosome release. Consequently, increased cellular accumulation of doxorubicin contributes to cytotoxicity and enhanced antitumor effects (87).

Furthermore, in agreement with previous studies, we confirmed that a combination of antihistamines with either doxorubicin and/or immune-checkpoint inhibition equips the host with strong immunologic memory (15, 18). The prominent immune response in animals receiving the combination treatment is attributed to the improved T-cell trafficking and particularly the increased ratio of effector CD8+ T-cell population to suppressive Tregs compared with the groups with no or limited memory response. Although the cytotoxic nature of doxorubicin has been reported to boost T-cell immunity, here we did not observe any alteration in T-cell infiltration in either tumor model. In K7M2, however, we found that doxorubicin synergizes with immunotherapy to promote CD8+ T-cell proliferation and T-cell attachment to vessels, independently of hypoxia alleviation, suggesting that other mechanisms may underline such antitumor responses. Consistent with previous studies (62, 88), our findings support an IFNγ-dependent mechanism by which ketotifen facilitates vascular normalization and, subsequently, increases perfusion, which in turn upregulates adhesion molecules necessary to support T-cell extravasation, increases recruitment, and doubles the fraction of CD8+ T cells proliferating in these tumors, while decreasing the polarization of the immature myeloid cell population into MDSCs (89–92). MDSCs concentrate in hypoxic cores and form a potent barrier to tumor immunity (71, 93–95). Besides MDSCs, M2-like macrophages have been associated with immunosuppression. It has been previously described that the inhibition of the histamine receptor H1 on M2-like macrophages blocks the membrane translocation of immune-checkpoint VISTA, thereby revitalizing T-cell cytotoxic function and restoring response to anti–PD-1 treatment (82). Nevertheless, further research including studies in MC-deficient mice is required to conclude whether these immunologic responses are MC- or ketotifen-driven.

Finally, we explored the translational possibility of monitoring tissue elastic properties using ultrasound SWE as a noninvasive and reliable biomarker of responsiveness to immune-checkpoint inhibition. Like our studies in murine breast tumor models (12, 18, 96), we show here that the reduction of tumor elastic modulus increases the efficacy of immunotherapy and chemotherapy in sarcoma tumors, highlighting further the potential of tumor stiffness to serve as a predictive marker of response. Future studies assessing perfusion status during the course of treatment using contrast-enhanced ultrasound could be used to test the hypothesis that mechanical biomarkers can be developed to predict the response of tumors to immunotherapies and chemotherapies. Ultrasound SWE is already used in the clinic for the evaluation of liver fibrosis and has been investigated in patients and animal models with various cancer types as a marker of response to chemotherapy (97–99).

Our findings from the clinical evaluation of ketotifen in sarcoma patients further demonstrate that measuring and monitoring tumor stiffness using noninvasive, clinically applied ultrasound SWE can be used as a predictive biomarker, particularly for tumors with a low incidence rate and of different histologic subtypes, such as sarcomas. Stabilizing MCs holds great promise for overcoming resistance to immunotherapy in solid tumors, which are associated with high levels of MCs or their products and have a poor prognosis, and warrants further investigation.

In conclusion, in this study, we primarily describe a previously unexplored interaction between tumor-resident MCs and CAFs and demonstrate the ability of the approved antihistamine and mast cell stabilizer, ketotifen, to restore patho-physiologic abnormalities of the TME. As cancer might be considered a TME disease, we show that pretreatment with ketotifen establishes favorable conditions within the TME toward potentiating chemoimmunotherapy and establishing durable immune responses in murine models of sarcoma.

J.D. Martin reports personal fees from NanoCarrier Co., Ltd outside the submitted work. In addition, J.D. Martin has a patent for cancer treatment using ketotifen in combination with a checkpoint inhibitor pending and licensed to Materia Therapeutics Inc and a patent for cancer treatment using bosentan in combination with a checkpoint inhibitor pending and licensed to Materia Therapeutics Inc; Dr. Martin owns equity in Materia Therapeutics Inc. T. Stylianopoulos reports a patent for cancer treatment using ketotifen in combination with a checkpoint inhibitor pending and licensed to Materia Therapeutics Inc. No disclosures were reported by the other authors.

M. Panagi: Conceptualization, resources, investigation, methodology, writing–original draft. F. Mpekris: Investigation, methodology. C. Voutouri: Data curation, software, formal analysis. A.G. Hadjigeorgiou: Data curation, software, formal analysis. C. Symeonidou: Validation, visualization, methodology. E. Porfyriou: Project administration. C. Michael: Resources, investigation, methodology. A. Stylianou: Formal analysis, investigation. J.D. Martin: Conceptualization, writing–review and editing. H. Cabral: Conceptualization, supervision. A. Constantinidou: Conceptualization, supervision. T. Stylianopoulos: Conceptualization, supervision, funding acquisition, writing–review and editing.

We thank the patients and all the investigators who participated in these studies. The authors gratefully recognize the contributions of staff from the Bank of Cyprus Oncology Center in supporting the centralized screening of patients. The authors were fully responsible for all content and editorial decisions, were involved at all stages of manuscript development, and approved the final version.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Jenkins
L
,
Jungwirth
U
,
Avgustinova
A
,
Iravani
M
,
Mills
A
,
Haider
S
, et al
.
Cancer-associated fibroblasts suppress CD8 T-cell infiltration and confer resistance to immune-checkpoint blockade
.
Cancer Res
2022
;
16
:
2904
17
.
2.
Van der Leun
AM
,
Thommen
DS
,
Schumacher
TN
.
CD8 T cell states in human cancer: insights from single-cell analysis
.
Nat Rev Cancer
2020
;
20
:
218
32
.
3.
Panagi
M
,
Pilavaki
P
,
Constantinidou
A
,
Stylianopoulos
T
.
Immunotherapy in soft tissue and bone sarcoma: unraveling the barriers to effectiveness
.
Theranostics
2022
;
12
:
6106
29
.
4.
Pilavaki
P
,
Panagi
M
,
Arifi
S
,
Jones
RL
,
Stylianopoulos
T
,
Constantinidou
A
.
Exploring the landscape of immunotherapy approaches in sarcomas
.
Front Oncol
2022
;
12
:
1069963
.
5.
Stubbs
M
,
McSheehy
PM
,
Griffiths
JR
,
Bashford
CL
.
Causes and consequences of tumour acidity and implications for treatment
.
Mol Med Today
2000
;
6
:
15
9
.
6.
Chauhan
VP
,
Boucher
Y
,
Ferrone
CR
,
Roberge
S
,
Martin
JD
,
Stylianopoulos
T
, et al
.
Compression of pancreatic tumor blood vessels by hyaluronan is caused by solid stress and not interstitial fluid pressure
.
Cancer Cell
2014
;
26
:
14
5
.
7.
Stylianopoulos
T
,
Martin
JD
,
Snuderl
M
,
Mpekris
F
,
Jain
SR
,
Jain
RK
.
Coevolution of solid stress and interstitial fluid pressure in tumors during progression: implications for vascular collapse
.
Cancer Res
2013
;
73
:
3833
41
.
8.
Vavourakis
V
,
Wijeratne
PA
,
Shipley
R
,
Loizidou
M
,
Stylianopoulos
T
,
Hawkes
DJ
.
A validated multiscale in-silico model for mechano-sensitive tumour angiogenesis and growth
.
PLoS Comput Biol
2017
;
13
:
e1005259
.
9.
Angeli
S
,
Stylianopoulos
T
.
Biphasic modeling of brain tumor biomechanics and response to radiation treatment
.
J Biomech
2016
;
49
:
1524
31
.
10.
Chauhan
VP
,
Martin
JD
,
Liu
H
,
Lacorre
DA
,
Jain
SR
,
Kozin
SV
, et al
.
Angiotensin inhibition enhances drug delivery and potentiates chemotherapy by decompressing tumour blood vessels
.
Nat Commun
2013
;
4
:
1
11
.
11.
Huang
Y
,
Goel
S
,
Duda
DG
,
Fukumura
D
,
Jain
RK
.
Vascular normalization as an emerging strategy to enhance cancer immunotherapy
.
Cancer Res
2013
;
73
:
2943
8
.
12.
Voutouri
C
,
Panagi
M
,
Mpekris
F
,
Stylianou
A
,
Michael
C
,
Averkiou
MA
, et al
.
Endothelin inhibition potentiates cancer immunotherapy revealing mechanical biomarkers predictive of response
.
Adv Ther
2021
:
2000289
.
13.
Barsoum
IB
,
Smallwood
CA
,
Siemens
DR
,
Graham
CH
.
A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells
.
Cancer Res
2014
;
74
:
665
74
.
14.
Panagi
M
,
Voutouri
C
,
Mpekris
F
,
Papageorgis
P
,
Martin
MR
,
Martin
JD
, et al
.
TGF-β inhibition combined with cytotoxic nanomedicine normalizes triple negative breast cancer microenvironment towards anti-tumor immunity
.
Theranostics
2020
;
10
:
1910
.
15.
Mpekris
F
,
Panagi
M
,
Voutouri
C
,
Martin
JD
,
Samuel
R
,
Takahashi
S
, et al
.
Normalizing the microenvironment overcomes vessel compression and resistance to nano-immunotherapy in breast cancer lung metastasis
.
Adv Sci
2021
;
8
:
2001917
.
16.
Martin
JD
,
Panagi
M
,
Wang
C
,
Khan
TT
,
Martin
MR
,
Voutouri
C
, et al
.
Dexamethasone increases cisplatin-loaded nanocarrier delivery and efficacy in metastatic breast cancer by normalizing the tumor microenvironment
.
ACS Nano
2019
;
13
:
6396
408
.
17.
Polydorou
C
,
Mpekris
F
,
Papageorgis
P
,
Voutouri
C
,
Stylianopoulos
T
.
Pirfenidone normalizes the tumor microenvironment to improve chemotherapy
.
Oncotarget
2017
;
8
:
24506
.
18.
Panagi
M
,
Mpekris
F
,
Chen
P
,
Voutouri
C
,
Nakagawa
Y
,
Martin
JD
, et al
.
Polymeric micelles effectively reprogram the tumor microenvironment to potentiate nano-immunotherapy in mouse breast cancer models
.
Nat Commun
2022
;
13
:
1
14
.
19.
Papageorgis
P
,
Polydorou
C
,
Mpekris
F
,
Voutouri
C
,
Agathokleous
E
,
Kapnissi-Christodoulou
CP
, et al
.
Tranilast-induced stress alleviation in solid tumors improves the efficacy of chemo-and nanotherapeutics in a size-independent manner
.
Sci Rep
2017
;
7
:
46140
.
20.
Diop-Frimpong
B
,
Chauhan
VP
,
Krane
S
,
Boucher
Y
,
Jain
RK
.
Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors
.
Proc Natl Acad Sci
2011
;
108
:
2909
14
.
21.
Zhao
Y
,
Cao
J
,
Melamed
A
,
Worley
M
,
Gockley
A
,
Jones
D
, et al
.
Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma
.
Proc Natl Acad Sci
2019
;
116
:
2210
9
.
22.
Kalkusova
K
,
Smite
S
,
Darras
E
,
Taborska
P
,
Stakheev
D
,
Vannucci
L
, et al
.
Mast cells and dendritic cells as cellular immune checkpoints in immunotherapy of solid tumors
.
Int J Mol Sci
2022
;
23
:
11080
.
23.
Visciano
C
,
Liotti
F
,
Prevete
N
,
Cali
G
,
Franco
R
,
Collina
F
, et al
.
Mast cells induce epithelial-to-mesenchymal transition and stem cell features in human thyroid cancer cells through an IL-8–Akt–Slug pathway
.
Oncogene
2015
;
34
:
5175
86
.
24.
Ma
Y
,
Hwang
RF
,
Logsdon
CD
,
Ullrich
SE
.
Dynamic mast cell–stromal cell interactions promote growth of pancreatic cancer
.
Cancer Res
2013
;
73
:
3927
37
.
25.
Dabiri
S
,
Huntsman
D
,
Makretsov
N
,
Cheang
M
,
Gilks
B
,
Badjik
C
, et al
.
The presence of stromal mast cells identifies a subset of invasive breast cancers with a favorable prognosis
.
Mod Pathol
2004
;
17
:
690
5
.
26.
Varricchi
G
,
Galdiero
MR
,
Loffredo
S
,
Marone
G
,
Iannone
R
,
Marone
G
, et al
.
Are mast cells MASTers in cancer?
Front Immunol
2017
;
8
:
424
.
27.
Ribatti
D
,
Vacca
A
,
Ria
R
,
Marzullo
A
,
Nico
B
,
Filotico
R
, et al
.
Neovascularisation, expression of fibroblast growth factor-2, and mast cells with tryptase activity increase simultaneously with pathological progression in human malignant melanoma
.
Eur J Cancer
2003
;
39
:
666
74
.
28.
Mangia
A
,
Malfettone
A
,
Rossi
R
,
Paradiso
A
,
Ranieri
G
,
Simone
G
, et al
.
Tissue remodelling in breast cancer: human mast cell tryptase as an initiator of myofibroblast differentiation
.
Histopathol
2011
;
58
:
1096
106
.
29.
Coussens
LM
,
Raymond
WW
,
Bergers
G
,
Laig-Webster
M
,
Behrendtsen
O
,
Werb
Z
, et al
.
Inflammatory mast cells up-regulate angiogenesis during squamous epithelial carcinogenesis
.
Genes Dev
1999
;
13
:
1382
97
.
30.
Esposito
I
,
Menicagli
M
,
Funel
N
,
Bergmann
F
,
Boggi
U
,
Mosca
F
, et al
.
Inflammatory cells contribute to the generation of an angiogenic phenotype in pancreatic ductal adenocarcinoma
.
J Clin Pathol
2004
;
57
:
630
6
.
31.
Baram
D
,
Vaday
GG
,
Salamon
P
,
Drucker
I
,
Hershkoviz
R
,
Mekori
YA
.
Human mast cells release metalloproteinase-9 on contact with activated T cells: juxtacrine regulation by TNF-α
.
J Immunol
2001
;
167
:
4008
16
.
32.
Gulliksson
M
,
Carvalho
RF
,
Ullerås
E
,
Nilsson
G
.
Mast cell survival and mediator secretion in response to hypoxia
.
PLoS One
2010
;
5
:
e12360
.
33.
Walczak-Drzewiecka
A
,
Ratajewski
M
,
Wagner
W
,
Dastych
J
.
HIF-1α is up-regulated in activated mast cells by a process that involves calcineurin and NFAT
.
J Immunol
2008
;
181
:
1665
72
.
34.
Huang
Y
,
Yuan
J
,
Righi
E
,
Kamoun
WS
,
Ancukiewicz
M
,
Nezivar
J
, et al
.
Vascular normalizing doses of antiangiogenic treatment reprogram the immunosuppressive tumor microenvironment and enhance immunotherapy
.
Proc Natl Acad Sci
2012
;
109
:
17561
6
.
35.
Hatfield
SM
,
Kjaergaard
J
,
Lukashev
D
,
Schreiber
TH
,
Belikoff
B
,
Abbott
R
, et al
.
Immunological mechanisms of the antitumor effects of supplemental oxygenation
.
Sci Transl Med
2015
;
7
:
277ra30
.
36.
Jayaprakash
P
,
Ai
M
,
Liu
A
,
Budhani
P
,
Bartkowiak
T
,
Sheng
J
, et al
.
Targeted hypoxia reduction restores T cell infiltration and sensitizes prostate cancer to immunotherapy
.
J Clin Invest
2018
;
128
:
5137
49
.
37.
Murugesan
G
,
Weigle
B
,
Crocker
PR
.
Siglec and anti-siglec therapies
.
Curr Opin Chem Biol
2021
;
62
:
34
42
.
38.
Abbaspour Babaei
M
,
Kamalidehghan
B
,
Saleem
M
,
Huri
HZ
,
Ahmadipour
F
.
Receptor tyrosine kinase (c-kit) inhibitors: a potential therapeutic target in cancer cells
.
Drug Des Devel Ther
2016
:
2443
59
.
39.
Somasundaram
R
,
Connelly
T
,
Choi
R
,
Choi
H
,
Samarkina
A
,
Li
L
, et al
.
Tumor-infiltrating mast cells are associated with resistance to anti-PD-1 therapy
.
Nat Commun
2021
;
12
:
346
.
40.
Abe
M1
,
Kurosawa
M
,
Ishikawa
O
,
Miyachi
Y
,
Kido
H
.
Mast cell tryptase stimulates both human dermal fibroblast proliferation and type I collagen production
.
Clin Exp Allergy
1998
;
28
:
1509
17
.
41.
Woodman
L
,
Siddiqui
S
,
Cruse
G
,
Sutcliffe
A
,
Saunders
R
,
Kaur
D
, et al
.
Mast cells promote airway smooth muscle cell differentiation via autocrine up-regulation of TGF-β1
.
J Immunol
2008
;
181
:
5001
7
.
42.
Mariathasan
S
,
Turley
SJ
,
Nickles
D
,
Castiglioni
A
,
Yuen
K
,
Wang
Y
, et al
.
TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells
.
Nature
2018
;
554
:
544
8
.
43.
Grisez
BT
,
Ray
JJ
,
Bostian
PA
,
Markel
JE
,
Lindsey
BA
.
Highly metastatic K7M2 cell line: a novel murine model capable of in vivo imaging via luciferase vector transfection
.
J Orthop Res
2018
;
36
:
2296
304
.
44.
Abe
M
,
Kurosawa
M
,
Ishikawa
O
,
Miyachi
Y
.
Effect of mast cell–derived mediators and mast cell–related neutral proteases on human dermal fibroblast proliferation and type I collagen production
.
J Allergy Clin Immunol
2000
;
106
:
S78
84
.
45.
Ruoss
SJ
,
Hartmann
T
,
Caughey
GH
.
Mast cell tryptase is a mitogen for cultured fibroblasts
.
J Clin Invest
1991
;
88
:
493
9
.
46.
Staats
HF
,
Kirwan
SM
,
Choi
HW
,
Shelburne
CP
,
Abraham
SN
,
Leung
GY
, et al
.
A mast cell degranulation screening assay for the identification of novel mast cell activating agents
.
Med Chem Comm
2013
;
4
:
88
94
.
47.
McNeil
BD
,
Pundir
P
,
Meeker
S
,
Han
L
,
Undem
BJ
,
Kulka
M
, et al
.
Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions
.
Nature
2015
;
519
:
237
41
.
48.
Border
WA
,
Noble
NA
.
Transforming growth factor β in tissue fibrosis
.
N Engl J Med
1994
;
331
:
1286
92
.
49.
Monument
MJ
,
Hart
DA
,
Befus
AD
,
Salo
PT
,
Zhang
M
,
Hildebrand
KA
.
The mast cell stabilizer ketotifen fumarate lessens contracture severity and myofibroblast hyperplasia: a study of a rabbit model of posttraumatic joint contractures
.
J Bone Joint Surg Am
2010
;
92
:
1468
.
50.
Zhang
Y
,
Berger
SA
.
Ketotifen reverses MDR1-mediated multidrug resistance in human breast cancer cells in vitro and alleviates cardiotoxicity induced by doxorubicin in vivo
.
Cancer Chemother Pharmacol
2003
;
51
:
407
14
.
51.
Baba
A
,
Tachi
M
,
Ejima
Y
,
Endo
Y
,
Toyama
H
,
Matsubara
M
, et al
.
Anti-allergic drugs tranilast and ketotifen dose-dependently exert mast cell-stabilizing properties
.
Cell Physiol Biochem
2016
;
38
:
15
27
.
52.
Burks
CA
,
Rhodes
SD
,
Bessler
WK
,
Chen
S
,
Smith
A
,
Gehlhausen
JR
, et al
.
Ketotifen modulates mast cell chemotaxis to kit-ligand, but does not impact mast cell numbers, degranulation, or tumor behavior in neurofibromas of Nf1-deficient mice
.
Mol Cancer Ther
2019
;
18
:
2321
30
.
53.
Stylianou
A
,
Mpekris
F
,
Voutouri
C
,
Papoui
A
,
Constantinidou
A
,
Kitiris
E
, et al
.
Nanomechanical properties of solid tumors as treatment monitoring biomarkers
.
Acta Biomater
2022
;
154
:
324
34
.
54.
Stylianopoulos
T
.
The solid mechanics of cancer and strategies for improved therapy
.
J Biomech Eng
2017
;
139
:
021004
.
55.
Stylianopoulos
T
,
Martin
JD
,
Chauhan
VP
,
Jain
SR
,
Diop-Frimpong
B
,
Bardeesy
N
, et al
.
Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors
.
Proc Natl Acad Sci
2012
;
109
:
15101
8
.
56.
Voutouri
C
,
Polydorou
C
,
Papageorgis
P
,
Gkretsi
V
,
Stylianopoulos
T
.
Hyaluronan-derived swelling of solid tumors, the contribution of collagen and cancer cells, and implications for cancer therapy
.
Neoplasia
2016
;
18
:
732
41
.
57.
Voutouri
C
,
Stylianopoulos
T
.
Accumulation of mechanical forces in tumors is related to hyaluronan content and tissue stiffness
.
PLoS One
2018
;
13
:
e0193801
.
58.
Rozenberg
I
,
Sluka
SH
,
Rohrer
L
,
Hofmann
J
,
Becher
B
,
Akhmedov
A
, et al
.
Histamine H1 receptor promotes atherosclerotic lesion formation by increasing vascular permeability for low-density lipoproteins
.
Arterioscler Thromb Vasc Biol
2010
;
30
:
923
30
.
59.
Guo
M
,
Breslin
JW
,
Wu
MH
,
Gottardi
CJ
,
Yuan
SY
.
VE-cadherin and β-catenin binding dynamics during histamine-induced endothelial hyperpermeability
.
Am J Physiol Cell Physiol
2008
;
294
:
977
84
.
60.
Marks
RM
,
Roche
WR
,
Czerniecki
M
,
Penny
R
,
Nelson
DS
.
Mast cell granules cause proliferation of human microvascular endothelial cells
.
Lab Invest
1986
;
55
:
289
94
.
61.
Sörbo
J
,
Jakobsson
A
,
Norrby
K
.
Mast-cell histamine is angiogenic through receptors for histamine1 and histamine2
.
Int J Exp Pathol
1994
;
75
:
43
.
62.
Tian
L
,
Goldstein
A
,
Wang
H
,
Lo
HC
,
Kim
IS
,
Welte
T
, et al
.
Mutual regulation of tumour vessel normalization and immunostimulatory reprogramming
.
Nature
2017
;
544
:
250
4
.
63.
Mpekris
F
,
Voutouri
C
,
Baish
JW
,
Duda
DG
,
Munn
LL
,
Stylianopoulos
T
, et al
.
Combining microenvironment normalization strategies to improve cancer immunotherapy
.
Proc Natl Acad Sci
2020
;
117
:
3728
37
.
64.
Melo
V
,
Bremer
E
,
Martin
JD
.
Towards immunotherapy-induced normalization of the tumor microenvironment
.
Front Cell Dev Biol
2022
;
10
:
908389
.
65.
McLachlan
GJ
,
Lee
SX
,
Rathnayake
SI
.
Finite mixture models
.
Annu Rev Stat Appl
2019
;
6
:
355
78
.
66.
Benci
JL
,
Xu
B
,
Qiu
Y
,
Wu
TJ
,
Dada
H
,
Twyman-Saint Victor
C
, et al
.
Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade
.
Cell
2016
;
167
:
1540
,1554. e12.
67.
Rüegg
C
,
Yilmaz
A
,
Bieler
G
,
Bamat
J
,
Chaubert
P
,
Lejeune
FJ
.
Evidence for the involvement of endothelial cell integrin αVβ3 in the disruption of the tumor vasculature induced by TNF and IFN-γ
.
Nat Med
1998
;
4
:
408
14
.
68.
Meunier
M
,
Delisle
J
,
Bergeron
J
,
Rineau
V
,
Baron
C
,
Perreault
C
.
T cells targeted against a single minor histocompatibility antigen can cure solid tumors
.
Nat Med
2005
;
11
:
1222
9
.
69.
Shankaran
V
,
Ikeda
H
,
Bruce
AT
,
White
JM
,
Swanson
PE
,
Old
LJ
, et al
.
IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity
.
Nature
2001
;
410
:
1107
11
.
70.
Zheng
X
,
Fang
Z
,
Liu
X
,
Deng
S
,
Zhou
P
,
Wang
X
, et al
.
Increased vessel perfusion predicts the efficacy of immune checkpoint blockade
.
J Clin Invest
2018
;
128
:
2104
15
.
71.
Alizadeh
D
,
Trad
M
,
Hanke
NT
,
Larmonier
CB
,
Janikashvili
N
,
Bonnotte
B
, et al
.
Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer
.
Cancer Res
2014
;
74
:
104
18
.
72.
Wang
Z
,
Till
B
,
Gao
Q
.
Chemotherapeutic agent-mediated elimination of myeloid-derived suppressor cells
.
Oncoimmunology
2017
;
6
:
e1331807
.
73.
Segura-Villalobos
D
,
Ramírez-Moreno
IG
,
Martínez-Aguilar
M
,
Ibarra-Sánchez
A
,
Muñoz-Bello
JO
,
Anaya-Rubio
I
, et al
.
Mast cell–tumor interactions: Molecular mechanisms of recruitment, intratumoral communication and potential therapeutic targets for tumor growth
.
Cells
2022
;
11
:
349
.
74.
Akers
IA
,
Parsons
M
,
Hill
MR
,
Hollenberg
MD
,
Sanjar
S
,
Laurent
GJ
, et al
.
Mast cell tryptase stimulates human lung fibroblast proliferation via protease-activated receptor-2
.
Am J Physiol Lung Cell Mol Physiol
2000
;
278
:
L193
201
.
75.
Murray
LA
,
Argentieri
RL
,
Farrell
FX
,
Bracht
M
,
Sheng
H
,
Whitaker
B
, et al
.
Hyper-responsiveness of IPF/UIP fibroblasts: Interplay between TGFβ1, IL-13 and CCL2
.
Int J Biochem Cell Biol
2008
;
40
:
2174
82
.
76.
Kim
DK
,
Jeong
J
,
Lee
DS
,
Hyeon
DY
,
Park
GW
,
Jeon
S
, et al
.
PD-L1-directed PlGF/VEGF blockade synergizes with chemotherapy by targeting CD141 cancer-associated fibroblasts in pancreatic cancer
.
Nat Commun
2022
;
13
:
6292
.
77.
Hyeon
DY
,
Nam
D
,
Han
Y
,
Kim
DK
,
Kim
G
,
Kim
D
, et al
.
Proteogenomic landscape of human pancreatic ductal adenocarcinoma in an Asian population reveals tumor cell-enriched and immune-rich subtypes
.
Nat Cancer
2023
;
4
:
290
307
.
78.
Chitty
JL
,
Yam
M
,
Perryman
L
,
Parker
AL
,
Skhinas
JN
,
Setargew
YF
, et al
.
A first-in-class pan-lysyl oxidase inhibitor impairs stromal remodeling and enhances gemcitabine response and survival in pancreatic cancer
.
Nat Cancer
2023
:
1
19
.
79.
Murphy
JE
,
Wo
JY
,
Ryan
DP
,
Clark
JW
,
Jiang
W
,
Yeap
BY
, et al
.
Total neoadjuvant therapy with FOLFIRINOX in combination with losartan followed by chemoradiotherapy for locally advanced pancreatic cancer: a phase 2 clinical trial
.
JAMA Oncol
2019
;
5
:
1020
7
.
80.
Boucher
Y
,
Posada
JM
,
Subudhi
S
,
Kumar
AS
,
Rosario
SR
,
Gu
L
, et al
.
Addition of losartan to FOLFIRINOX and chemoradiation reduces immunosuppression-associated genes, Tregs, and FOXP3 cancer cells in locally advanced pancreatic cancer
.
Clin Cancer Res
2023
;
29
:
1605
19
.
81.
Lin
Y
,
Wang
H
,
Tsai
M
,
Su
Y
,
Yang
M
,
Chien
C
.
Angiotensin II receptor blockers valsartan and losartan improve survival rate clinically and suppress tumor growth via apoptosis related to PI3K/AKT signaling in nasopharyngeal carcinoma
.
Cancer
2021
;
127
:
1606
19
.
82.
Li
H
,
Xiao
Y
,
Li
Q
,
Yao
J
,
Yuan
X
,
Zhang
Y
, et al
.
The allergy mediator histamine confers resistance to immunotherapy in cancer patients via activation of the macrophage histamine receptor H1
.
Cancer Cell
2022
;
40
:
36,52. e9
.
83.
Fritz
I
,
Wagner
P
,
Olsson
H
.
Improved survival in several cancers with use of H1-antihistamines desloratadine and loratadine
.
Transl Oncol
2021
;
14
:
101029
.
84.
Zou
W
,
Wolchok
JD
,
Chen
L
.
PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: mechanisms, response biomarkers, and combinations
.
Sci Transl Med
2016
;
8
:
328rv4
.
85.
Topalian
SL
,
Taube
JM
,
Anders
RA
,
Pardoll
DM
.
Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy
.
Nat Rev Cancer
2016
;
16
:
275
87
.
86.
Sharma
P
,
Allison
JP
.
The future of immune checkpoint therapy
.
Science
2015
;
348
:
56
61
.
87.
Khan
FM
,
Saleh
E
,
Alawadhi
H
,
Harati
R
,
Zimmermann
W
,
El-Awady
R
.
Inhibition of exosome release by ketotifen enhances sensitivity of cancer cells to doxorubicin
.
Cancer Biol Ther
2018
;
19
:
25
33
.
88.
De Palma
M
,
Jain
RK
.
CD4 T cell activation and vascular normalization: two sides of the same coin?
Immunity
2017
;
46
:
773
5
.
89.
Becker
JC
,
Andersen
MH
,
Schrama
D
,
thor Straten
P
.
Immune-suppressive properties of the tumor microenvironment
.
Cancer Immunol Immunother
2013
;
62
:
1137
48
.
90.
Ben-Shoshan
J
,
Maysel-Auslender
S
,
Mor
A
,
Keren
G
,
George
J
.
Hypoxia controls CD4 CD25 regulatory T-cell homeostasis via hypoxia-inducible factor-1α
.
Eur J Immunol
2008
;
38
:
2412
8
.
91.
Corzo
CA
,
Condamine
T
,
Lu
L
,
Cotter
MJ
,
Youn
J
,
Cheng
P
, et al
.
HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment
.
J Exp Med
2010
;
207
:
2439
53
.
92.
Quintana
FJ
,
Basso
AS
,
Iglesias
AH
,
Korn
T
,
Farez
MF
,
Bettelli
E
, et al
.
Control of Treg and TH 17 cell differentiation by the aryl hydrocarbon receptor
.
Nature
2008
;
453
:
65
71
.
93.
Zheng
B
,
Ren
T
,
Huang
Y
,
Guo
W
.
Apatinib inhibits migration and invasion as well as PD-L1 expression in osteosarcoma by targeting STAT3
.
Biochem Biophys Res Commun
2018
;
495
:
1695
701
.
94.
Ribatti
D
,
Ranieri
G
,
Nico
B
,
Benagiano
V
,
Crivellato
E
.
Tryptase and chymase are angiogenic in vivo in the chorioallantoic membrane assay
.
Int J Dev Biol
2011
;
55
:
99
102
.
95.
Lv
Y
,
Zhao
Y
,
Wang
X
,
Chen
N
,
Mao
F
,
Teng
Y
, et al
.
Increased intratumoral mast cells foster immune suppression and gastric cancer progression through TNF-α-PD-L1 pathway
.
J Immunother Cancer
2019
;
7
:
1
15
.
96.
Voutouri
C
,
Mpekris
F
,
Panagi
M
,
Krolak
C
,
Michael
C
,
Martin
JD
, et al
.
Ultrasound stiffness and perfusion markers correlate with tumor volume responses to immunotherapy
.
Acta Biomater
2023
;
167
:
121
34
.
97.
Kwon
JH
,
Yoo
SH
,
Nam
SW
,
Lee
YJ
,
Shin
YR
.
Diagnostic role of contrast-enhanced ultrasound in the discrimination of malignant portal vein thrombosis in patients with hepatocellular carcinoma
.
Anticancer Res
2020
;
40
:
4351
63
.
98.
Evans
A
,
Armstrong
S
,
Whelehan
P
,
Thomson
K
,
Rauchhaus
P
,
Purdie
C
, et al
.
Can shear-wave elastography predict response to neoadjuvant chemotherapy in women with invasive breast cancer?
Br J Cancer
2013
;
109
:
2798
802
.
99.
Mpekris
F
,
Panagi
M
,
Michael
C
,
Voutouri
C
,
Tsuchiya
M
,
Wagatsuma
C
, et al
.
Translational nanomedicine potentiates immunotherapy in sarcoma by normalizing the microenvironment
.
J Controlled Release
2023
;
353
:
956
64
.
This open access article is distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0) license.