1
|
Charalambous A, Mpekris F, Panagi M, Voutouri C, Michael C, Gabizon AA, Stylianopoulos T. Tumor Microenvironment Reprogramming Improves Nanomedicine-Based Chemo-Immunotherapy in Sarcomas. Mol Cancer Ther 2024; 23:1555-1567. [PMID: 38940284 DOI: 10.1158/1535-7163.mct-23-0772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/16/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024]
Abstract
Sarcomas are a heterogeneous group of rare cancers that originate in soft tissues or bones. Their complexity and tendency for metastases make treatment challenging, highlighting the need for new therapeutic approaches to improve patient survival. The difficulties in treating these cancers primarily stem from abnormalities within the tumor microenvironment (TME), which leads to reduced blood flow and oxygen levels in tumors. Consequently, this hampers the effective delivery of drugs to tumors and diminishes treatment efficacy despite higher toxic doses of chemotherapy. In this study, we tested the mechanotherapeutic ketotifen combined with either pegylated liposomal doxorubicin (PLD) or pegylated liposomal coencapsulated alendronate-doxorubicin (PLAD) plus anti-programmed cell death protein 1 antibody in mouse models of fibrosarcoma and osteosarcoma. We found that ketotifen successfully reprogrammed the TME by reducing tumor stiffness and increasing perfusion, proven by changes measured by shear-wave elastography and contrast-enhanced ultrasound, respectively, and enhanced the therapeutic efficacy of our nanomedicine-based chemo-immunotherapy protocols. Furthermore, we observed a trend toward improved antitumor responses when nano-chemotherapy is given alongside anti-programmed cell death protein 1 and when the immunomodulator alendronate was present in the treatment. We next investigated the mechanisms of action of this combination. Ketotifen combined with nanomedicine-based chemo-immunotherapy increased T-cell infiltration, specifically cytotoxic CD8+ T cells and CD4+ T helper cells, and decreased the number of regulatory T cells. In addition, the combination also altered the polarization of tumor-associated macrophages, favoring the M1 immune-supportive phenotype over the M2 immunosuppressive phenotype. Collectively, our findings provide evidence that ketotifen-induced TME reprogramming can improve the efficacy of nanomedicine-based chemo-immunotherapy in sarcomas.
Collapse
Affiliation(s)
- Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Alberto A Gabizon
- Nano-Oncology Research Center, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
2
|
Voutouri C, Englezos D, Zamboglou C, Strouthos I, Papanastasiou G, Stylianopoulos T. A convolutional attention model for predicting response to chemo-immunotherapy from ultrasound elastography in mouse tumor models. COMMUNICATIONS MEDICINE 2024; 4:203. [PMID: 39420199 PMCID: PMC11487255 DOI: 10.1038/s43856-024-00634-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND In the era of personalized cancer treatment, understanding the intrinsic heterogeneity of tumors is crucial. Despite some patients responding favorably to a particular treatment, others may not benefit, leading to the varied efficacy observed in standard therapies. This study focuses on the prediction of tumor response to chemo-immunotherapy, exploring the potential of tumor mechanics and medical imaging as predictive biomarkers. We have extensively studied "desmoplastic" tumors, characterized by a dense and very stiff stroma, which presents a substantial challenge for treatment. The increased stiffness of such tumors can be restored through pharmacological intervention with mechanotherapeutics. METHODS We developed a deep learning methodology based on shear wave elastography (SWE) images, which involved a convolutional neural network (CNN) model enhanced with attention modules. The model was developed and evaluated as a predictive biomarker in the setting of detecting responsive, stable, and non-responsive tumors to chemotherapy, immunotherapy, or the combination, following mechanotherapeutics administration. A dataset of 1365 SWE images was obtained from 630 tumors from our previous experiments and used to train and successfully evaluate our methodology. SWE in combination with deep learning models, has demonstrated promising results in disease diagnosis and tumor classification but their potential for predicting tumor response prior to therapy is not yet fully realized. RESULTS We present strong evidence that integrating SWE-derived biomarkers with automatic tumor segmentation algorithms enables accurate tumor detection and prediction of therapeutic outcomes. CONCLUSIONS This approach can enhance personalized cancer treatment by providing non-invasive, reliable predictions of therapeutic outcomes.
Collapse
Affiliation(s)
- Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Demetris Englezos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University of Freiburg - Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Giorgos Papanastasiou
- Archimedes Unit, Athena Research Centre, Athens, Greece
- School of Computer Science and Electronic Engineering University of Essex, Wivenhoe Park, UK
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
3
|
Wang Z, Miao F, Gu L, Zhang R, Ma Y, Li Y, Zheng J, Lin Z, Gao Y, Huang L, Shen Y, Wu T, Luo F, Li W. Stimulator of Interferon Genes-Activated Biomimetic Dendritic Cell Nanovaccine as a Chemotherapeutic Booster to Enhance Systemic Fibrosarcoma Treatment. ACS NANO 2024; 18:24219-24235. [PMID: 39172516 DOI: 10.1021/acsnano.4c05657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Fibrosarcoma, a malignant mesenchymal tumor, is characterized by aggressive invasiveness and a high recurrence rate, leading to poor prognosis. Anthracycline drugs, such as doxorubicin (DOX), represent the frontline chemotherapy for fibrosarcoma, but often exhibit suboptimal efficacy. Recently, exploiting the stimulator of interferon genes (STING)-mediated innate immunity has emerged as a hopeful strategy for cancer treatment. Integrating chemotherapy with immunomodulators in chemo-immunotherapy has shown potential for enhancing treatment outcomes. Herein, we introduce an advanced dendritic cell (DC) nanovaccine, cGAMP@PLGA@CRTM (GP@CRTM), combined with low-dose DOX to enhance fibrosarcoma chemo-immunotherapy. The nanovaccine consists of poly(lactic-co-glycolic acid) (PLGA) nanoparticles encapsulating the STING agonist 2,3-cGAMP (cGAMP@PLGA, GP) as its core, and a calreticulin (CRT) high-expressing fibrosarcoma cell membrane (CRTM) as the shell. Exposing CRT on the vaccine surface aids in recruiting DCs and stimulating uptake, facilitating efficient simultaneous delivery of STING agonists and tumor antigens to DCs. This dual delivery method effectively activates the STING pathway in DCs, triggering sustained immune stimulation. Simultaneously, low-dose DOX reduces chemotherapy-related side effects, directly kills a subset of tumor cells, and increases tumor immunogenicity, thus further amplifying immune therapeutic performance. Hence, these findings demonstrate the potential of DC nanovaccine GP@CRTM as a booster for chemotherapy. Synergistically combining low-dose DOX with the DC nanovaccine emerges as a powerful chemo-immunotherapy strategy, optimizing systemic fibrosarcoma therapy.
Collapse
Affiliation(s)
- Zhao Wang
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Fenglin Miao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Lingwei Gu
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Ruyi Zhang
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Yuan Ma
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Ying Li
- Heji Hospital Affiliated with Changzhi Medical College, Changzhi 046000, Shanxi, China
| | - Jialiang Zheng
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Zhenhang Lin
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Yilai Gao
- Cancer Research Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361000, Fujian, China
| | - Liyong Huang
- Fuzhou No. 1 Hospital Affiliated with Fujian Medical University, Fuzhou 350000, Fu Jian, China
| | - Ye Shen
- Shanghai Jiangxia Blood Technology Co., Ltd. Shanghai 200000, China
| | - Ting Wu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Fanghong Luo
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Wengang Li
- Department of Hepatobiliary Surgery, Xiang'an Hospital of Xiamen University, Cancer Research Center, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| |
Collapse
|
4
|
Serras A, Faustino C, Pinheiro L. Functionalized Polymeric Micelles for Targeted Cancer Therapy: Steps from Conceptualization to Clinical Trials. Pharmaceutics 2024; 16:1047. [PMID: 39204392 PMCID: PMC11359152 DOI: 10.3390/pharmaceutics16081047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is still ranked among the top three causes of death in the 30- to 69-year-old age group in most countries and carries considerable societal and macroeconomic costs that differ depending on the cancer type, geography, and patient gender. Despite advances in several pharmacological approaches, the lack of stability and specificity, dose-related toxicity, and limited bioavailability of chemotherapy (standard therapy) pose major obstacles in cancer treatment, with multidrug resistance being a driving factor in chemotherapy failure. The past three decades have been the stage for intense research activity on the topic of nanomedicine, which has resulted in many nanotherapeutics with reduced toxicity, increased bioavailability, and improved pharmacokinetics and therapeutic efficacy employing smart drug delivery systems (SDDSs). Polymeric micelles (PMs) have become an auspicious DDS for medicinal compounds, being used to encapsulate hydrophobic drugs that also exhibit substantial toxicity. Through preclinical animal testing, PMs improved pharmacokinetic profiles and increased efficacy, resulting in a higher safety profile for therapeutic drugs. This review focuses on PMs that are already in clinical trials, traveling the pathways from preclinical to clinical studies until introduction to the market.
Collapse
Affiliation(s)
| | - Célia Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa (ULisboa), Avenida Professor Gama PintoGama Pinto, 1649-003 Lisboa, Portugal; (A.S.); (L.P.)
| | | |
Collapse
|
5
|
Mpekris F, Panagi M, Charalambous A, Voutouri C, Stylianopoulos T. Modulating cancer mechanopathology to restore vascular function and enhance immunotherapy. Cell Rep Med 2024; 5:101626. [PMID: 38944037 PMCID: PMC11293360 DOI: 10.1016/j.xcrm.2024.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/12/2024] [Accepted: 06/07/2024] [Indexed: 07/01/2024]
Abstract
Solid tumor pathology, characterized by abnormalities in the tumor microenvironment (TME), challenges therapeutic effectiveness. Mechanical factors, including increased tumor stiffness and accumulation of intratumoral forces, can determine the success of cancer treatments, defining the tumor's "mechanopathology" profile. These abnormalities cause extensive vascular compression, leading to hypoperfusion and hypoxia. Hypoperfusion hinders drug delivery, while hypoxia creates an unfavorable TME, promoting tumor progression through immunosuppression, heightened metastatic potential, drug resistance, and chaotic angiogenesis. Strategies targeting TME mechanopathology, such as vascular and stroma normalization, hold promise in enhancing cancer therapies with some already advancing to the clinic. Normalization can be achieved using anti-angiogenic agents, mechanotherapeutics, immune checkpoint inhibitors, engineered bacterial therapeutics, metronomic nanomedicine, and ultrasound sonopermeation. Here, we review the methods developed to rectify tumor mechanopathology, which have even led to cures in preclinical models, and discuss their bench-to-bedside translation, including the derivation of biomarkers from tumor mechanopathology for personalized therapy.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
6
|
Panagi M, Mpekris F, Voutouri C, Hadjigeorgiou AG, Symeonidou C, Porfyriou E, Michael C, Stylianou A, Martin JD, Cabral H, Constantinidou A, Stylianopoulos T. Stabilizing Tumor-Resident Mast Cells Restores T-Cell Infiltration and Sensitizes Sarcomas to PD-L1 Inhibition. Clin Cancer Res 2024; 30:2582-2597. [PMID: 38578281 PMCID: PMC11145177 DOI: 10.1158/1078-0432.ccr-24-0246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/10/2024] [Accepted: 04/03/2024] [Indexed: 04/06/2024]
Abstract
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.
Collapse
Affiliation(s)
- Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G. Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | | | | | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas Stylianou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
- Basic and Translational Cancer Research Center, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | | | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Anastasia Constantinidou
- Bank of Cyprus Oncology Centre, Nicosia, Cyprus
- Cyprus Cancer Research Institute, Nicosia, Cyprus
- Medical School, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
7
|
Englezos D, Voutouri C, Stylianopoulos T. Machine learning analysis reveals tumor stiffness and hypoperfusion as biomarkers predictive of cancer treatment efficacy. Transl Oncol 2024; 44:101944. [PMID: 38552284 PMCID: PMC10990740 DOI: 10.1016/j.tranon.2024.101944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/06/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024] Open
Abstract
In the pursuit of advancing cancer therapy, this study explores the predictive power of machine learning in analyzing tumor characteristics, specifically focusing on the effects of tumor stiffness and perfusion (i.e., blood flow) on treatment efficacy. Recent advancements in oncology have highlighted the significance of these physiological properties of the tumor microenvironment in determining treatment outcomes. We delve into the relationship between these tumor attributes and the effectiveness of cancer therapies in preclinical tumor models. Utilizing robust statistical methods and machine learning algorithms, our research analyzes data from 1365 cases of various cancer types, assessing how tumor stiffness and perfusion influence the efficacy of treatment protocols. We also investigate the synergistic potential of combining drugs that modulate tumor stiffness and perfusion with standard cytotoxic treatments. By incorporating these predictors into treatment planning, our study aims to enhance the precision of cancer therapy, tailoring treatment to individual tumor profiles. Our findings demonstrate a significant correlation between stiffness/perfusion and treatment efficacy, highlighting a new way for personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Demetris Englezos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
8
|
Harkos C, Stylianopoulos T. Investigating the synergistic effects of immunotherapy and normalization treatment in modulating tumor microenvironment and enhancing treatment efficacy. J Theor Biol 2024; 583:111768. [PMID: 38401748 DOI: 10.1016/j.jtbi.2024.111768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
We developed a comprehensive mathematical model of cancer immunotherapy that takes into account: i) Immune checkpoint blockers (ICBs) and the interactions between cancer cells and the immune system, ii) characteristics of the tumor microenvironment, such as the tumor hydraulic conductivity, interstitial fluid pressure, and vascular permeability, iii) spatial and temporal variations of the modeled components within the tumor and the surrounding host tissue, iv) the transport of modeled components through the vasculature and between the tumor-host tissue with convection and diffusion, and v) modeling of the tumor draining lymph nodes were the antigen presentation and the development of cytotoxic immune cells take place. Our model successfully reproduced experimental data from various murine tumor types and predicted immune system profiling, which is challenging to achieve experimentally. It showed that combination of ICB therapy and normalization treatments, that aim to improve tumor perfusion, decreases interstitial fluid pressure and increases the concentration of both innate and adaptive immune cells at the tumor center rather than the periphery. Furthermore, using the model, we investigated the impact of modeled components on treatment outcomes. The analysis found that the number of functional vessels inside the tumor region and the ICB dose administered have the largest impact on treatment outcomes.
Collapse
Affiliation(s)
- Constantinos Harkos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
9
|
Mpekris F, Papaphilippou PC, Panagi M, Voutouri C, Michael C, Charalambous A, Marinov Dinev M, Katsioloudi A, Prokopi-Demetriades M, Anayiotos A, Cabral H, Krasia-Christoforou T, Stylianopoulos T. Pirfenidone-Loaded Polymeric Micelles as an Effective Mechanotherapeutic to Potentiate Immunotherapy in Mouse Tumor Models. ACS NANO 2023; 17:24654-24667. [PMID: 38054429 PMCID: PMC10753878 DOI: 10.1021/acsnano.3c03305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023]
Abstract
Ongoing research is actively exploring the use of immune checkpoint inhibitors to treat solid tumors by inhibiting the PD-1/PD-L1 axis and reactivating the function of cytotoxic T effector cells. Many types of solid tumors, however, are characterized by a dense and stiff stroma and are difficult to treat. Mechanotherapeutics have formed a recent class of drugs that aim to restore biomechanical abnormalities of the tumor microenvironment, related to increased stiffness and hypo-perfusion. Here, we have developed a polymeric formulation containing pirfenidone, which has been successful in restoring the tumor microenvironment in breast tumors and sarcomas. We found that the micellar formulation can induce similar mechanotherapeutic effects to mouse models of 4T1 and E0771 triple negative breast tumors and MCA205 fibrosarcoma tumors but with a dose 100-fold lower than that of the free pirfenidone. Importantly, a combination of pirfenidone-loaded micelles with immune checkpoint inhibition significantly delayed primary tumor growth, leading to a significant improvement in overall survival and in a complete cure for the E0771 tumor model. Furthermore, the combination treatment increased CD4+ and CD8+ T cell infiltration and suppressed myeloid-derived suppressor cells, creating favorable immunostimulatory conditions, which led to immunological memory. Ultrasound shear wave elastography (SWE) was able to monitor changes in tumor stiffness during treatment, suggesting optimal treatment conditions. Micellar encapsulation is a promising strategy for mechanotherapeutics, and imaging methods, such as SWE, can assist their clinical translation.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| | - Petri Ch. Papaphilippou
- Polymers
and Polymer Processing Laboratories, Department of Mechanical and
Manufacturing Engineering, University of
Cyprus, 1678 Nicosia, Cyprus
| | - Myrofora Panagi
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| | - Christina Michael
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| | - Mariyan Marinov Dinev
- Polymers
and Polymer Processing Laboratories, Department of Mechanical and
Manufacturing Engineering, University of
Cyprus, 1678 Nicosia, Cyprus
| | | | - Marianna Prokopi-Demetriades
- Theramir
Ltd, R&D Laboratory, 4101 Limassol, Cyprus
- Biomechanics
and Living Systems Analysis Laboratory, Cyprus University of Technology, 3036 Limassol, Cyprus
| | - Andreas Anayiotos
- Biomechanics
and Living Systems Analysis Laboratory, Cyprus University of Technology, 3036 Limassol, Cyprus
| | - Horacio Cabral
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, 113-8656 Tokyo, Japan
| | - Theodora Krasia-Christoforou
- Polymers
and Polymer Processing Laboratories, Department of Mechanical and
Manufacturing Engineering, University of
Cyprus, 1678 Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer
Biophysics Laboratory, Department of Mechanical and Manufacturing
Engineering, University of Cyprus, 1678 Nicosia, Cyprus
| |
Collapse
|
10
|
Voutouri C, Mpekris F, Panagi M, Krolak C, Michael C, Martin JD, Averkiou MA, Stylianopoulos T. Ultrasound stiffness and perfusion markers correlate with tumor volume responses to immunotherapy. Acta Biomater 2023:S1742-7061(23)00332-X. [PMID: 37321529 DOI: 10.1016/j.actbio.2023.06.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
Immunotherapy has revolutionized the treatment of dozens of cancers and became a standard of care for some tumor types. However, the majority of patients do not benefit from current immunotherapeutics and many develop severe toxicities. Therefore, the identification of biomarkers to classify patients as likely responders or non-responders to immunotherapy is a timely task. Here, we test ultrasound imaging markers of tumor stiffness and perfusion. Ultrasound imaging is non-invasive and clinically available and can be used both for stiffness and perfusion evaluation. In this study, we employed syngeneic orthotopic models of two breast cancers, a fibrosarcoma and melanoma, to demonstrate that ultrasound-derived measures of tumor stiffness and perfusion (i.e., blood volume) correlate with the efficacy of immune checkpoint inhibition (ICI) in terms of changes in primary tumor volume. To modulate tumor stiffness and perfusion and thus, get a range of therapeutic outcomes, we employed the mechanotherapeutic tranilast. Mechanotherapeutics combined with ICI are advancing through clinical trials, but biomarkers of response have not been tested until now. We found the existence of linear correlations between tumor stiffness and perfusion imaging biomarkers as well as strong linear correlations between the stiffness and perfusion markers with ICI efficacy on primary tumor growth rates. Our findings set the basis for ultrasound imaging biomarkers predictive of ICI therapy in combination with mechanotherapeutics. STATEMENT OF SIGNIFICANCE: Hypothesis: Monitoring Tumor Microenvironment (TME) mechanical abnormalities can predict the efficacy of immune checkpoint inhibition (ICI) and provide biomarkers predictive of response. Tumor stiffening and solid stress elevation are hallmarks of tumor patho-physiology in desmoplastic tumors. They induce hypo-perfusion and hypoxia by compressing tumor vessels, posing major barriers to immunotherapy. Mechanotherapeutics is a new class of drugs that target the TME to reduce stiffness and improve perfusion and oxygenation. In this study, we show that measures of stiffness and perfusion derived from ultrasound shear wave elastography and contrast enhanced ultrasound can provide biomarkers of tumor response.
Collapse
Affiliation(s)
- Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus
| | - Connor Krolak
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Christina Michael
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus
| | | | | | | |
Collapse
|
11
|
Kou Q, Huang Y, Su Y, Lu L, Li X, Jiang H, Huang R, Li J, Nie X. Erythrocyte membrane-camouflaged DNA-functionalized upconversion nanoparticles for tumor-targeted chemotherapy and immunotherapy. NANOSCALE 2023. [PMID: 37161583 DOI: 10.1039/d3nr00542a] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
A synergistic combination of treatment with immunogenic cell death (ICD) inducers and immunoadjuvants may be a practical way to boost the anticancer response and successfully induce an immune response. The use of HR@UCNPs/CpG-Apt/DOX, new biomimetic drug delivery nanoparticles generated to combat breast cancer, is reported here as a unique strategy to produce immunogenicity and boost cancer immunotherapy. HR@UCNPs/CpG-Apt/DOX (HR-UCAD) consists of two parts. The core is composed of an immunoadjuvant CpG (a toll-like receptor 9 agonist) fused with a dendritic cell-specific aptamer sequence (CpG-Apt) to decorate upconversion nanoparticles (UCNPs) with the successful intercalation of doxorubicin (DOX) into the consecutive base pairs of Apt-CpG to construct an immune nanodrug UCNPs@CpG-Apt/DOX. The targeting molecule hyaluronic acid (HA) was inserted into a red blood cell membrane (RBCm) to form the shell (HR). HR-UCAD possessed a strong capacity to specifically induce ICD. Following DOX-induced ICD of cancer cells, sufficient exposure to tumor antigens and UCNPs@CpG-Apt (UCA) activated the tumor-specific immune response and reversed the immunosuppressive tumor microenvironment. In addition, HR-UCAD has good biocompatibility and increases the active tumor-targeting effect. Furthermore, HR-UCAD exhibits excellent near-infrared upconversion luminescence emission at 804 nm under irradiation with a 980 nm laser, which has great potential in biomedical imaging. Thus, the RBCm-camouflaged drug delivery system is a promising targeted chemotherapy and immunotherapy nanocomplex that could be used for effective targeted breast cancer treatment.
Collapse
Affiliation(s)
- Qinjie Kou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yufen Huang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yanrong Su
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lu Lu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Xisheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Haiye Jiang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| | - Xinmin Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- Hunan Engineering Technology Research Center of Optoelectronic Health Detection, Changsha, 410000, Hunan, China.
| |
Collapse
|