1
|
Wehrenberg-Klee E, Hampilos P, Austin EE, Ataeinia B, MacPherson A, LaSalle T, Mahmood U. Evaluating the Impact of Adjunctive Partial Cryoablation on Dual Checkpoint Inhibitor Immunotherapy Response in a Murine Model. Radiol Imaging Cancer 2024; 6:e230187. [PMID: 39485112 PMCID: PMC11615628 DOI: 10.1148/rycan.230187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 11/03/2024]
Abstract
Purpose To evaluate the impact of adjunctive partial cryoablation on checkpoint inhibitor (CPI) immunotherapy response. Materials and Methods One hundred fifty-six mice (equal number of male and female animals) with dual-implanted tumor models were treated with dual CPI or a vehicle and randomized to treatment of a single tumor with partial cryoablation. Tumors were followed for 60 days following cryoablation for response assessment. In additional groups, the tumor microenvironment was characterized via flow cytometry, cytokine analysis, and immunohistochemistry. Statistical comparison was made between the different treatment groups regarding T-cell infiltration and activation characteristics within the noncryoablated tumor and cytokine levels within the partially ablated tumor. Additionally, qualitative assessment of T-cell activation within the cryoablated and noncryoablated tumors at immunofluorescence was carried out. Results At 60 days following treatment, CPI and adjunctive cryoablation-treated MC-38 mice had a significantly increased survival rate (79%) compared with mice treated with CPI alone (61%; P < .001). CT-26 mice also had an increased survival rate (57% vs 35%, respectively; P = .04). Following cryoablation, increases in inflammatory cytokines and chemokines within the treated tumors were observed. Flow cytometry of noncryoablated tumor showed increased CD8 T-cell activation. Immunofluorescence and histologic evaluation following cryoablation further demonstrated a robust CD8 T-cell and myeloid infiltrate. Conclusion Adjunctive cryoablation significantly increased the response to dual CPI in multiple cancer models at both partially ablated and distant (nonablated) tumor sites. Immune analysis suggests cryoablation promotes a vigorous immune response within the partially cryoablated tumor that increases activation of the adaptive immune system within distant tumor sites. Keywords: Cancer, Cryoablation, Checkpoint Inhibitor Immunotherapy, Tumor Response Supplemental material is available for this article. © RSNA, 2024.
Collapse
Affiliation(s)
- Eric Wehrenberg-Klee
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Perry Hampilos
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Emily E. Austin
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Bahar Ataeinia
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Abigail MacPherson
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Thomas LaSalle
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| | - Umar Mahmood
- From the Department of Radiology, Center for Precision Imaging,
Martinos Center for Biomedical Imaging, Massachusetts General Hospital,
149 13th St, Rm 5.407, Charlestown, MA 02129 (E.W.K., P.H., E.E.A., B.A.,
A.M., T.L., U.M.); and Department of Radiology, Division of Interventional
Radiology, Massachusetts General Hospital, Boston, Mass (E.W.K., P.H.)
| |
Collapse
|
2
|
Hansen SB, Unal B, Kuzu OF, Saatcioglu F. Immunological facets of prostate cancer and the potential of immune checkpoint inhibition in disease management. Theranostics 2024; 14:6913-6934. [PMID: 39629128 PMCID: PMC11610136 DOI: 10.7150/thno.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 12/06/2024] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men and a major cause of cancer-related deaths. Whereas localized PCa can be cured by surgery and radiotherapy, metastatic disease can be treated, but is not curable. Inhibition of androgen signaling remains the main therapeutic intervention for treatment of metastatic PCa, in addition to chemotherapy, radionuclide therapy and emerging targeted therapies. Although initial responses are favorable, resistance to these therapies invariably arise with development of castration resistant PCa (CRPC) and lethal phenotypes. Recent findings have implicated the crosstalk between PCa cells and the tumor microenvironment (TME) as a key factor for disease progression and metastasis, and the immune system is becoming an increasingly attractive target for therapy. Given the striking success of immune checkpoint inhibitors (ICIs) in various cancer types, preclinical and clinical studies have begun to explore their potential in PCa. It has become clear that the PCa TME is largely immunosuppressive, and ICI therapy does not have efficacy for PCa. Intense effort is therefore being made in the field to understand the mechanisms of suppression and to turn the immunosuppressive TME into an immune active one that would enable ICI efficacy. Herein we examine this recent body of knowledge and how the mutational landscape of PCa integrates with an immunosuppressive TME to circumvent ICI-mediated T-cell activity and tumor killing. We then review the emerging potential success of combinatorial ICI approaches, utility of careful patient selection, and potential novel strategies to improve the efficacy of ICI for PCa therapy.
Collapse
Affiliation(s)
| | - Bilal Unal
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Omer Faruk Kuzu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
3
|
Liu D, Wang L, Guo Y. Advances in and prospects of immunotherapy for prostate cancer. Cancer Lett 2024; 601:217155. [PMID: 39127338 DOI: 10.1016/j.canlet.2024.217155] [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: 05/30/2024] [Revised: 07/07/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
Immunotherapy has shown promising therapeutic effects in hematological malignancies and certain solid tumors and has emerged as a critical and highly potential treatment modality for cancer. However, prostate cancer falls under the category of immune-resistant cold tumors, for which immunotherapy exhibits limited efficacy in patients with solid tumors. Thus, it is important to gain a deeper understanding of the tumor microenvironment in prostate cancer to facilitate immune system activation and overcome immune suppression to advance immunotherapy for prostate cancer. In this review, we discuss the immunosuppressive microenvironment of prostate cancer, which is characterized by the presence of few tumor-infiltrating lymphocytes, abundant immunosuppressive cells, low immunogenicity, and a noninflammatory phenotype, which significantly influences the efficacy of immunotherapy for prostate cancer. Immunotherapy is mainly achieved by activating the host immune system and overcoming immunosuppression. In this regard, we summarize the therapeutic advances in immune checkpoint blockade, immunogenic cell death, reversal of the immunosuppressive tumor microenvironment, tumor vaccines, immune adjuvants, chimeric antigen receptor T-cell therapy, and overcoming penetration barriers in prostate cancer, with the aim of providing novel research insights and approaches to enhance the effectiveness of immunotherapy for prostate cancer.
Collapse
Affiliation(s)
- Deng Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China; Bioengineering College, Chongqing University, Chongqing, 400044, China
| | - Luofu Wang
- Department of Urology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Yanli Guo
- Department of Ultrasound, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
4
|
Su X, Li J, Xu X, Ye Y, Wang C, Pang G, Liu W, Liu A, Zhao C, Hao X. Strategies to enhance the therapeutic efficacy of anti-PD-1 antibody, anti-PD-L1 antibody and anti-CTLA-4 antibody in cancer therapy. J Transl Med 2024; 22:751. [PMID: 39123227 PMCID: PMC11316358 DOI: 10.1186/s12967-024-05552-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Although immune checkpoint inhibitors (anti-PD-1 antibody, anti-PD-L1 antibody, and anti-CTLA-4 antibody) have displayed considerable success in the treatment of malignant tumors, the therapeutic effect is still unsatisfactory for a portion of patients. Therefore, it is imperative to develop strategies to enhance the effect of these ICIs. Increasing evidence strongly suggests that the key to this issue is to transform the tumor immune microenvironment from a state of no or low immune infiltration to a state of high immune infiltration and enhance the tumor cell-killing effect of T cells. Therefore, some combination strategies have been proposed and this review appraise a summary of 39 strategies aiming at enhancing the effectiveness of ICIs, which comprise combining 10 clinical approaches and 29 foundational research strategies. Moreover, this review improves the comprehensive understanding of combination therapy with ICIs and inspires novel ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Su
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Jian Li
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiao Xu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Youbao Ye
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Cailiu Wang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Guanglong Pang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Wenxiu Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Ang Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Changchun Zhao
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, 730000, China
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, No. 204 Donggang West Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
5
|
Gu J, Yu Z, Tang X, Chen W, Deng X, Zhu X. Cryoablation combined with dual immune checkpoint blockade enhances antitumor efficacy in hepatocellular carcinoma model mice. Int J Hyperthermia 2024; 41:2373319. [PMID: 38955354 DOI: 10.1080/02656736.2024.2373319] [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: 04/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Cryoablation (Cryo) is a minimally invasive treatment for tumors. Cryo can activate the body's immune response, although it is typically weak. The immune response induced by Cryo in hepatocellular carcinoma (HCC) is poorly understood. PD-1 and CTLA-4 monoclonal antibodies are immune checkpoint inhibitors used in immunotherapy for tumors. The combined use of these antibodies with Cryo may enhance the immune effect. METHODS A Balb/c mouse model of HCC was established and treated with Cryo, immune checkpoint blockade (ICB), or Cryo + ICB (combination therapy). The growth trend of right untreated tumors and survival time of mice were determined. The expression of apoptosis-related proteins was detected by Western blot (WB) assay. The percentages of immune cells and immunosuppressive cells were analyzed by flow cytometry. The numbers of infiltrating T lymphocytes were checked by immunohistochemistry, and the levels of T-cell-associated cytokines were detected by Quantitative real-time Polymerase Chain Reaction (qRT-PCR) assays and Enzyme-Linked Immunosorbent Assays (ELISA) assays. RESULTS Cryo + ICB inhibited the growth of right untreated tumors, promoted tumor cell apoptosis, and prolonged the survival time of mice. Local T-cell infiltration in right tumor tissues increased after the combination therapy, while the number of immunosuppressive cells was significantly reduced. In addition, the combination therapy may induce the production of multiple Th1-type cytokines but reduce the production of Th2-type cytokines. CONCLUSIONS Cryo can activate CD8+ and CD4+ T-cell immune responses. Cryo + ICB can relieve the immunosuppressive tumor microenvironment and shift the Th1/Th2 balance toward Th1 dominance, further enhancing the Cryo-induced T-cell immune response and resulting in a stronger antitumor immune response.
Collapse
Affiliation(s)
- Jun Gu
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Zepeng Yu
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xiangxiang Tang
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, China
| | - Wenying Chen
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xuedong Deng
- Center for Medical Ultrasound, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Xiaoli Zhu
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
6
|
Séguier D, Adams ES, Kotamarti S, D'Anniballe V, Michael ZD, Deivasigamani S, Olivier J, Villers A, Hoimes C, Polascik TJ. Intratumoural immunotherapy plus focal thermal ablation for localized prostate cancer. Nat Rev Urol 2024; 21:290-302. [PMID: 38114768 DOI: 10.1038/s41585-023-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 12/21/2023]
Abstract
Major advances have been made in the use of immunotherapy for the treatment of solid tumours, including the use of intratumourally injected immunotherapy instead of systemically delivered immunotherapy. The success of immunotherapy in prostate cancer treatment has been limited to specific populations with advanced disease, which is thought to be a result of prostate cancer being an immunologically 'cold' cancer. Accordingly, combining intratumoural immunotherapy with other treatments that would increase the immunological heat of prostate cancer is of interest. Thermal ablation therapy is currently one of the main strategies used for the treatment of localized prostate cancer and it causes immunological activation against prostate tissue. The use of intratumoural immunotherapy as an adjunct to thermal ablation offers the potential to elicit a systemic and lasting adaptive immune response to cancer-specific antigens, leading to a synergistic effect of combination therapy. The combination of thermal ablation and immunotherapy is currently in the early stages of investigation for the treatment of multiple solid tumour types, and the potential for this combination therapy to also offer benefit to prostate cancer patients is exciting.
Collapse
Affiliation(s)
- Denis Séguier
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA.
- Department of Urology, Lille University, Lille, France.
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Eric S Adams
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Srinath Kotamarti
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Vincent D'Anniballe
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Zoe D Michael
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Sriram Deivasigamani
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | - Jonathan Olivier
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Arnauld Villers
- Department of Urology, Lille University, Lille, France
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER; UMR9020-U1277), Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Christopher Hoimes
- Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, 27708, USA
| | - Thomas J Polascik
- Department of Surgery, Division of Urology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| |
Collapse
|
7
|
Liu Q, Zhang C, Chen X, Han Z. Modern cancer therapy: cryoablation meets immune checkpoint blockade. Front Oncol 2024; 14:1323070. [PMID: 38384806 PMCID: PMC10881233 DOI: 10.3389/fonc.2024.1323070] [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: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cryoablation, as a minimally invasive technology for the treatment of tumors, destroys target tumors with lethal low temperatures. It simultaneously releases a large number of tumor-specific antigens, pro-inflammatory cytokines, and nucleoproteins, known as "danger signals", activating the body's innate and adaptive immune responses. However, tumor cells can promote the inactivation of immune effector cells by reprogramming immune checkpoints, leading to the insufficiency of these antigens to induce an immune response capable of eradicating the tumor. Immune checkpoint blockers rejuvenate exhausted T cells by blocking immune checkpoints that induce programmed death of T cells, and are therefore considered a promising therapeutic strategy to enhance the immune effects of cryoablation. In this review, we provide a detailed explanation of the immunological mechanisms of cryoablation and articulate the theoretical basis and research progress of the treatment of cancer with cryoablation combined with immune checkpoint blockers. Preliminary data indicates that this combined treatment strategy exhibits good synergy and has been proven to be safe and effective.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Chunyang Zhang
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Xuxin Chen
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhihai Han
- Department of Pulmonary and Critical Care Medicine, The Sixth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Navy Clinical College, the Fifth School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
- College of Pulmonary and Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
8
|
Mustafa AR, Miyasato D, Wehrenberg-Klee E. Synergizing Thermal Ablation Modalities with Immunotherapy: Enough to Induce Systemic Antitumoral Immunity? J Vasc Interv Radiol 2024; 35:185-197. [PMID: 38272639 DOI: 10.1016/j.jvir.2023.10.033] [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: 07/01/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 01/27/2024] Open
Abstract
Thermal ablation modalities (cryoablation, radiofrequency ablation, and microwave ablation) have long been noted to occasionally induce a systemic antitumoral response. With the widespread use of checkpoint inhibitors, there is a significant interest in whether thermal ablation can promote immune system tumor recognition and increase checkpoint inhibitor response rates. In this review, we examine the current state of preclinical and clinical evidence examining the combination of checkpoint inhibitor therapies and thermal ablation modalities as well as discuss remaining the unanswered questions and directions for future research.
Collapse
Affiliation(s)
- Abdul Rehman Mustafa
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | | | - Eric Wehrenberg-Klee
- Division of Interventional Radiology, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
9
|
Gu C, Wang X, Wang K, Xie F, Chen L, Ji H, Sun J. Cryoablation triggers type I interferon-dependent antitumor immunity and potentiates immunotherapy efficacy in lung cancer. J Immunother Cancer 2024; 12:e008386. [PMID: 38272564 PMCID: PMC10824009 DOI: 10.1136/jitc-2023-008386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Cryoablation is a minimally invasive option for patients with medically inoperable non-small cell lung cancer (NSCLC) and can trigger abscopal immune-regulatory effects. However, it remains unclear how cryoablation affects the host-level immune response in NSCLC. In this study, we investigated the local and systemic immunological effects of cryoablation and the potential of combining cryoablation with programmed cell death protein 1 (PD-1) blockade to boost immunotherapy efficacy in NSCLC. METHODS We first investigated systemic immunological effects induced by cryoablation in patients with early-stage NSCLC. Subsequently, we explored cryoablation-induced antitumor immunity and the underlying biological mechanisms using KP (Kras G12D/+, Tp53 -/-) mutant lung cancer cell allograft mouse models. Moreover, the synergistic efficacy of cryoablation and PD-1 blockade was explored in both mouse models and patients with unresectable NSCLC. RESULTS We found that cryoablation significantly increased circulating CD8+ T cell subpopulations and proinflammatory cytokines in patients with early-stage NSCLC. In lung cancer cell allograft mouse models, we demonstrated that cryoablation resulted in abscopal growth inhibition of contralateral, non-ablated tumors. Integrated analysis of bulk, single-cell RNA and T cell receptor (TCR) sequencing data revealed that cryoablation reprogrammed the intratumoral immune microenvironment and increased CD8+ T cell infiltration with higher effector signature, interferon (IFN) response, and cytolytic activity. Mechanistically, cryoablation promoted antitumor effect through the STING-dependent type I IFN signaling pathway, and type I IFN signaling blockade attenuated this antitumor effect. We also found that the combination of PD-1 blockade with cryoablation further inhibited tumor growth compared with either treatment alone in an allograft mouse model. Moreover, the combination therapy induced notable tumor suppression and CD8+ T cell infiltration in patients with unresectable NSCLC. CONCLUSIONS Our results provide mechanistic insights into how cryoablation triggers the antitumor immune effect in lung cancer, thereby potentiating programmed cell death ligand 1 (PD-L1)/PD-1 blockade efficacy in the clinical treatment of NSCLC.
Collapse
Affiliation(s)
- Chuanjia Gu
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| | - Xue Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Kaiyu Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fangfang Xie
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, Guangdong, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Jiayuan Sun
- Department of Respiratory Endoscopy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Center of Respiratory Endoscopy, Shanghai, China
| |
Collapse
|
10
|
Yu ZP, Sun XW, He YP, Gu J, Jin Y. PD-1 monoclonal antibodies enhance the cryoablation-induced antitumor immune response: a breast cancer murine model research. Int J Hyperthermia 2023; 40:2164625. [PMID: 36966808 DOI: 10.1080/02656736.2022.2164625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND It has been demonstrated that cryoablation (Cryo) causes specific T-cell immune responses in the body; however, it is not sufficient to prevent tumor recurrence and metastasis. In this report, we evaluated changes in the tumor immune microenvironment (TIME) in distant tumor tissues after Cryo and investigated the immunosuppressive mechanisms that limit the efficacy of Cryo. METHODS Bilateral mammary tumor models were established in mice, and we first observed the dynamic changes in immune cells and cytokines at different time points after Cryo. Then, we confirmed that the upregulation of PD-1 and PD-L1 signaling in the contralateral tumor tissue was closely related to the immunosuppressive state in the TIME at the later stage after Cryo. Finally, we also evaluated the synergistic antitumor effects of Cryo combined with PD-1 monoclonal antibody (mAb) in the treatment of breast cancer (BC) mouse. RESULTS We found that Cryo can stimulate the body's immune response, but it also induces immunosuppression. The elevated PD-1/PD-L1 expression in distant tumor tissues at the later stage after Cryo was closely related to the immunosuppressive state in the TIME but also created the conditions for Cryo combined with PD-1 mAb for BC mouse treatment. Cryo + PD-1 mAb could improve the immunosuppressive state of tumors and enhance the Cryo-induced immune response, thus exerting a synergistic antitumor effect. CONCLUSIONS The PD-1/PD-L1 axis plays an important role in suppressing Cryo-induced antitumor immune responses. This study provides a theoretical basis for Cryo combined with PD-1 mAb therapy in clinical BC patients.
Collapse
|
11
|
He M, Cao Y, Chi C, Zhao J, Chong E, Chin KXC, Tan NZV, Dmitry K, Yang G, Yang X, Hu K, Enikeev M. Unleashing novel horizons in advanced prostate cancer treatment: investigating the potential of prostate specific membrane antigen-targeted nanomedicine-based combination therapy. Front Immunol 2023; 14:1265751. [PMID: 37795091 PMCID: PMC10545965 DOI: 10.3389/fimmu.2023.1265751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Prostate cancer (PCa) is a prevalent malignancy with increasing incidence in middle-aged and older men. Despite various treatment options, advanced metastatic PCa remains challenging with poor prognosis and limited effective therapies. Nanomedicine, with its targeted drug delivery capabilities, has emerged as a promising approach to enhance treatment efficacy and reduce adverse effects. Prostate-specific membrane antigen (PSMA) stands as one of the most distinctive and highly selective biomarkers for PCa, exhibiting robust expression in PCa cells. In this review, we explore the applications of PSMA-targeted nanomedicines in advanced PCa management. Our primary objective is to bridge the gap between cutting-edge nanomedicine research and clinical practice, making it accessible to the medical community. We discuss mainstream treatment strategies for advanced PCa, including chemotherapy, radiotherapy, and immunotherapy, in the context of PSMA-targeted nanomedicines. Additionally, we elucidate novel treatment concepts such as photodynamic and photothermal therapies, along with nano-theragnostics. We present the content in a clear and accessible manner, appealing to general physicians, including those with limited backgrounds in biochemistry and bioengineering. The review emphasizes the potential benefits of PSMA-targeted nanomedicines in enhancing treatment efficiency and improving patient outcomes. While the use of PSMA-targeted nano-drug delivery has demonstrated promising results, further investigation is required to comprehend the precise mechanisms of action, pharmacotoxicity, and long-term outcomes. By meticulous optimization of the combination of nanomedicines and PSMA ligands, a novel horizon of PSMA-targeted nanomedicine-based combination therapy could bring renewed hope for patients with advanced PCa.
Collapse
Affiliation(s)
- Mingze He
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Yu Cao
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Changliang Chi
- Department of Urology, First Hospital of Jilin University, Changchun, China
| | - Jiang Zhao
- Department of Urology, Xi’an First Hospital, Xi’an, China
| | - Eunice Chong
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Ke Xin Casey Chin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nicole Zian Vi Tan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Korolev Dmitry
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Guodong Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Xinyi Yang
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Kebang Hu
- Department of Urology, First Hospital of Jilin University, Changchun, China
| | - Mikhail Enikeev
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
12
|
He M, Cao Y, Chi C, Zhao J, Chong E, Chin KXC, Tan NZV, Dmitry K, Yang G, Yang X, Hu K, Enikeev M. Unleashing novel horizons in advanced prostate cancer treatment: investigating the potential of prostate specific membrane antigen-targeted nanomedicine-based combination therapy. Front Immunol 2023; 14. [DOI: https:/doi.org/10.3389/fimmu.2023.1265751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
Abstract
Prostate cancer (PCa) is a prevalent malignancy with increasing incidence in middle-aged and older men. Despite various treatment options, advanced metastatic PCa remains challenging with poor prognosis and limited effective therapies. Nanomedicine, with its targeted drug delivery capabilities, has emerged as a promising approach to enhance treatment efficacy and reduce adverse effects. Prostate-specific membrane antigen (PSMA) stands as one of the most distinctive and highly selective biomarkers for PCa, exhibiting robust expression in PCa cells. In this review, we explore the applications of PSMA-targeted nanomedicines in advanced PCa management. Our primary objective is to bridge the gap between cutting-edge nanomedicine research and clinical practice, making it accessible to the medical community. We discuss mainstream treatment strategies for advanced PCa, including chemotherapy, radiotherapy, and immunotherapy, in the context of PSMA-targeted nanomedicines. Additionally, we elucidate novel treatment concepts such as photodynamic and photothermal therapies, along with nano-theragnostics. We present the content in a clear and accessible manner, appealing to general physicians, including those with limited backgrounds in biochemistry and bioengineering. The review emphasizes the potential benefits of PSMA-targeted nanomedicines in enhancing treatment efficiency and improving patient outcomes. While the use of PSMA-targeted nano-drug delivery has demonstrated promising results, further investigation is required to comprehend the precise mechanisms of action, pharmacotoxicity, and long-term outcomes. By meticulous optimization of the combination of nanomedicines and PSMA ligands, a novel horizon of PSMA-targeted nanomedicine-based combination therapy could bring renewed hope for patients with advanced PCa.
Collapse
|
13
|
Lin M, Sun X, Lv L. New insights and options into the mechanisms and effects of combined targeted therapy and immunotherapy in prostate cancer. Mol Ther Oncolytics 2023; 29:91-106. [PMID: 37215386 PMCID: PMC10199166 DOI: 10.1016/j.omto.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Chronic inflammation is believed to drive prostate carcinogenesis by producing reactive oxygen species or reactive nitrogen species to induce DNA damage. This effect might subsequently cause epigenetic and genomic alterations, leading to malignant transformation. Although established therapeutic advances have extended overall survival, tumors in patients with advanced prostate cancer are prone to metastasis, transformation into metastatic castration-resistant prostate cancer, and therapeutic resistance. The tumor microenvironment (TME) of prostate cancer is involved in carcinogenesis, invasion and drug resistance. A plethora of preclinical studies have focused on immune-based therapies. Understanding the intricate TME system in prostate cancer may hold much promise for developing novel therapies, designing combinational therapeutic strategies, and further overcoming resistance to established treatments to improve the lives of prostate cancer patients. In this review, we discuss nonimmune components and various immune cells within the TME and their putative roles during prostate cancer initiation, progression, and metastasis. We also outline the updated fundamental research focusing on therapeutic advances of targeted therapy as well as combinational options for prostate cancer.
Collapse
Affiliation(s)
- Mingen Lin
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
| | - Xue Sun
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
| | - Lei Lv
- Nourse Centre for Pet Nutrition, Wuhu 241200, China
- Shanghai Chowsing Pet Products Co., Ltd, Shanghai 201103, China
| |
Collapse
|
14
|
Jiang M, Fiering S, Shao Q. Combining energy-based focal ablation and immune checkpoint inhibitors: preclinical research and clinical trials. Front Oncol 2023; 13:1153066. [PMID: 37251920 PMCID: PMC10211342 DOI: 10.3389/fonc.2023.1153066] [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: 01/28/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Energy-based focal therapy (FT) uses targeted, minimally invasive procedures to destroy tumors while preserving normal tissue and function. There is strong emerging interest in understanding how systemic immunity against the tumor can occur with cancer immunotherapy, most notably immune checkpoint inhibitors (ICI). The motivation for combining FT and ICI in cancer management relies on the synergy between the two different therapies: FT complements ICI by reducing tumor burden, increasing objective response rate, and reducing side effects of ICI; ICI supplements FT by reducing local recurrence, controlling distal metastases, and providing long-term protection. This combinatorial strategy has shown promising results in preclinical study (since 2004) and the clinical trials (since 2011). Understanding the synergy calls for understanding the physics and biology behind the two different therapies with distinctive mechanisms of action. In this review, we introduce different types of energy-based FT by covering the biophysics of tissue-energy interaction and present the immunomodulatory properties of FT. We discuss the basis of cancer immunotherapy with the emphasis on ICI. We examine the approaches researchers have been using and the results from both preclinical models and clinical trials from our exhaustive literature research. Finally, the challenges of the combinatory strategy and opportunities of future research is discussed extensively.
Collapse
Affiliation(s)
- Minhan Jiang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine and Dartmouth Health, Lebanon, NH, United States
| | - Qi Shao
- Department of Radiology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
15
|
Posa A, Contegiacomo A, Ponziani FR, Punzi E, Mazza G, Scrofani A, Pompili M, Goldberg SN, Natale L, Gasbarrini A, Sala E, Iezzi R. Interventional Oncology and Immuno-Oncology: Current Challenges and Future Trends. Int J Mol Sci 2023; 24:ijms24087344. [PMID: 37108507 PMCID: PMC10138371 DOI: 10.3390/ijms24087344] [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: 03/27/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Personalized cancer treatments help to deliver tailored and biologically driven therapies for cancer patients. Interventional oncology techniques are able to treat malignancies in a locoregional fashion, with a variety of mechanisms of action leading to tumor necrosis. Tumor destruction determines a great availability of tumor antigens that can be recognized by the immune system, potentially triggering an immune response. The advent of immunotherapy in cancer care, with the introduction of specific immune checkpoint inhibitors, has led to the investigation of the synergy of these drugs when used in combination with interventional oncology treatments. The aim of this paper is to review the most recent advances in the field of interventional oncology locoregional treatments and their interactions with immunotherapy.
Collapse
Affiliation(s)
- Alessandro Posa
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Andrea Contegiacomo
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Ernesto Punzi
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giulia Mazza
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Annarita Scrofani
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Maurizio Pompili
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Shraga Nahum Goldberg
- Division of Image-Guided Therapy, Department of Radiology, Hadassah Hebrew University Medical Center, Jerusalem 12000, Israel
| | - Luigi Natale
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Evis Sala
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| | - Roberto Iezzi
- Department of Diagnostic Imaging, Oncologic Radiotherapy and Hematology, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, L.go F. Vito 1, 00168 Rome, Italy
| |
Collapse
|
16
|
Chen Z, Meng L, Zhang J, Zhang X. Progress in the cryoablation and cryoimmunotherapy for tumor. Front Immunol 2023; 14:1094009. [PMID: 36761748 PMCID: PMC9907027 DOI: 10.3389/fimmu.2023.1094009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
With the rapid advancement of imaging equipment and minimally invasive technology, cryoablation technology is being used more frequently in minimally invasive treatment of tumors, primarily for patients with early tumors who voluntarily consent to ablation as well as those with advanced tumors that cannot be surgically removed or cannot be tolerated. Cryoablation is more effective and secure for target lesions than other thermal ablation methods like microwave and radiofrequency ablation (RFA). The study also discovered that cryoablation, in addition to causing tumor tissue necrosis and apoptosis, can facilitate the release of tumor-derived autoantigens into the bloodstream and activate the host immune system to elicit beneficial anti-tumor immunological responses against primary. This may result in regression of the primary tumor and distant metastasis. The additional effect called " Accompanying effects ". It is the basis of combined ablation and immunotherapy for tumor. At present, there is a lot of research on the mechanism of immune response induced by cryoablation. Trying to solve the question: how positively induce immune response. In this review, we focus on: 1. the immune effects induced by cryoablation. 2. the effect and mechanism of tumor immunotherapy combined with cryoablation. 3.The clinical research of this combination therapy in the treatment of tumors.
Collapse
Affiliation(s)
- Zenan Chen
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Liangliang Meng
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Radiology, Chinese People's Armed Police (PAP) Force Hospital of Beijing, Beijing, China
| | - Jing Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiao Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
17
|
Meng L, Zhang Z, Zhang X, Zhang X, Wei Y, Wu B, Xue X, Zhang X, He X, Xiao Y. Case report: Local cryoablation combined with pembrolizumab to eliminate lung metastases from ovarian clear cell carcinoma. Front Immunol 2022; 13:1006500. [DOI: 10.3389/fimmu.2022.1006500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
Ovarian clear cell carcinoma has a high recurrence rate with poor prognosis and is generally not sensitive to conventional platinum-based chemotherapy. Its less frequent occurrence of mutations such as BRCA limited the targeted therapies. Immunotherapy is not currently recommended as a first-line agent for ovarian cancer, and most patients are not yet able to benefit from it. Cryoablation can be used to treat solid systemic tumors, including ovarian cancer metastases, and can produce a limited anti-tumor immune response. The anti-tumor effects of cryoablation combined with immunotherapy have not been adequately confirmed. This study reports a case of a patient with ovarian clear cell carcinoma who underwent conventional adjuvant chemotherapy after initially surgical resection of the tumor. Unfortunately, cancer recurred and metastasized to the abdominal wall. After a series of painful chemotherapy and a second surgery, the cancer was still not effectively controlled, and the patient developed extensive metastases in the lung. The patient’s PD-L1 expression level also did not support solo immunotherapy. We pioneered the use of cryoablation to first eradicate the most significant lesion in the upper lobe of the left lung and then combined it with the PD-L1 inhibitor pembrolizumab to treat the patient with immunotherapy, which resulted in the complete eradication of the other multiple metastases in the lung and saved the patient’s life. Although the precise mechanism of action has not yet been explored, we have reason to believe that the combination of cryoablation and immune checkpoint inhibitor has a powerful synergistic anti-tumor effect, which is yet to be confirmed by more basic research and clinical applications in the next step.
Collapse
|
18
|
Dai Q, Cao B, Zhao S, Zhang A. Synergetic Thermal Therapy for Cancer: State-of-the-Art and the Future. Bioengineering (Basel) 2022; 9:bioengineering9090474. [PMID: 36135020 PMCID: PMC9495761 DOI: 10.3390/bioengineering9090474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
As a safe and minimal-invasive modality, thermal therapy has become an effective treatment in cancer treatment. Other than killing the tumor cells or destroying the tumor entirely, the thermal modality results in profound molecular, cellular and biological effects on both the targeted tissue, surrounding environments, and even the whole body, which has triggered the combination of the thermal therapy with other traditional therapies as chemotherapy and radiation therapy or new therapies like immunotherapy, gene therapy, etc. The combined treatments have shown encouraging therapeutic effects both in research and clinic. In this review, we have summarized the outcomes of the existing synergistic therapies, the underlying mechanisms that lead to these improvements, and the latest research in the past five years. Limitations and future directions of synergistic thermal therapy are also discussed.
Collapse
|
19
|
Li Y, Wang N, Zhao D, Wang J, Jiang L, Wang Y, Chen D, Wu Z, Zhou F, Yang Z. Cytoreductive prostate cryoablation and metronomic cyclophosphamide for metastatic hormone-sensitive prostate cancer. Future Oncol 2022; 18:2373-2380. [PMID: 35440168 DOI: 10.2217/fon-2021-1424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: This study reports the outcomes of cytoreductive prostate cryoablation and metronomic cyclophosphamide for the treatment of metastatic hormone-sensitive prostate cancer (mHSPC). Methods: Patients with mHSPC from the authors' prostate cancer database who had received cytoreductive prostate cryoablation and metronomic cyclophosphamide were identified retrospectively. Results: Eight consecutive patients were enrolled in the study. All the patients tolerated combination therapy. The median metastatic castration-resistant prostate cancer-free survival was 62.5 months. Seven patients (87.5%) had a prostate-specific antigen nadir <0.1 ng/ml. Dysuria and hematuria before prostate cryoablation disappeared within 1 month after cryosurgery, and no incontinence was seen after prostate cryoablation. No local therapy was needed during follow-up. Conclusion: Cytoreductive prostate cryoablation and metronomic cyclophosphamide prove an effective and safe combination therapy for mHSPC.
Collapse
Affiliation(s)
- Yonghong Li
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ning Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Diwei Zhao
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lijuan Jiang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yanjun Wang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Dong Chen
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhiming Wu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Fangjian Zhou
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhenyu Yang
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.,State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| |
Collapse
|
20
|
Jiao M, Zhang F, Teng W, Zhou C. MYBL2 is a Novel Independent Prognostic Biomarker and Correlated with Immune Infiltrates in Prostate Cancer. Int J Gen Med 2022; 15:3003-3030. [PMID: 35313552 PMCID: PMC8934167 DOI: 10.2147/ijgm.s351638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/17/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose MYB proto-oncogene like 2 (MYBL2) is a member of the MYB family of transcription factor genes and overexpressed in many cancers. We investigated the role of MYBL2 in the malignant progression of prostate cancer (PCa) and its relationship with immune infiltrates in PCa. Methods Gene expression level, clinicopathological parameters, Gene Ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway, tumor immune infiltration analysis were based on The Cancer Genome Atlas (TCGA) dataset. Gene set enrichment analysis (GSEA) and single-sample gene set enrichment analysis (ssGSEA) were conducted to analyze the correlation between MYBL2 and immune infiltrates. The data processing analysis based on R language. The relationship between MYBL2 expression and immune response in PCa was analyzed on TIMER 2.0. Results MYBL2 was overexpressed in PCa patients, and correlated with T-stage, Gleason score, primary therapy outcome and progress free interval (PFI) event. The multivariate Cox regression analysis revealed MYBL2 was an independent risk factor for PFI (HR=1.250, 95% CI=1.016–1.537, p=0.035). The receiver operating characteristic (ROC) curve for MYBL2 (AUC=0.887) and nomogram also confirmed the diagnostic value of MYBL2 in the treatment of PCa patients. Based on mRNA expression of MYBL2, PCa patients were divided into MYBL2-high group and MYBL2-low group, and analysis of MYBL2 associated KEGG and GO pathways using R language revealed that 6 immune-related signaling pathways were enriched in MYBL2-high expression phenotype. GSEA analysis showed that 3 hallmark gene sets related to immune response were significantly enriched in MYBL2-high group, ssGSEA analysis found that MYBL2 expression correlated with the expression of many tumor immune lymphocytes (CD8+T cells, neutrophil cells, macrophage cells and so on) and immune check point inhibitors (CD276, BTLA, TNFRSF18, HAVCR2 and CD70). Conclusion MYBL2 is a novel independent prognostic biomarker and MYBL2 may play a crucial role in tumor immune microenvironment of PCa.
Collapse
Affiliation(s)
- Meng Jiao
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
| | - Facai Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, People’s Republic of China
| | - Wei Teng
- College of Business, Gachon University, Seongnam, 13120, Republic of Korea
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People’s Republic of China
- Correspondence: Chengjun Zhou, Department of Pathology, The Second Hospital of Shandong University, 247 Beiyuan Street, Jinan, Shandong, 250033, People’s Republic of China, Email
| |
Collapse
|
21
|
Rangamuwa K, Leong T, Weeden C, Asselin-Labat ML, Bozinovski S, Christie M, John T, Antippa P, Irving L, Steinfort D. Thermal ablation in non-small cell lung cancer: a review of treatment modalities and the evidence for combination with immune checkpoint inhibitors. Transl Lung Cancer Res 2021; 10:2842-2857. [PMID: 34295682 PMCID: PMC8264311 DOI: 10.21037/tlcr-20-1075] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide, with approximately 1.6 million cancer related deaths each year. Prognosis is best in patients with early stage disease, though even then five-year survival is only 55% in some groups. Median survival for advanced non-small cell lung cancer (NSCLC) is 8–12 months with conventional treatment. Immune checkpoint inhibitor (ICI) therapy has revolutionised the treatment of NSCLC with significant long-term improvements in survival demonstrated in some patients with advanced NSCLC. However, only a small proportion of patients respond to ICI, suggesting the need for further techniques to harness the potential of ICI therapy. Thermal ablation utilizes the extremes of temperature to cause tumour destruction. Commonly used modalities are radiofrequency ablation (RFA), cryoablation and microwave ablation (MWA). At present thermal ablation is reserved for curative-intent therapy in patients with localized NSCLC who are unable to undergo surgical resection or stereotactic ablative body radiotherapy (SABR). Limited evidence suggests that thermal ablative modalities can upregulate an anticancer immune response in NSCLC. It is postulated that thermal ablation can increase tumour antigen release, which would initiate and upregulated steps in the cancer immunity cycle required to elicit an anticancer immune response. This article will review the current thermal ablative techniques and their ability to modulate an anti-cancer immune response with a view of using thermal ablation in conjunction with ICI therapy.
Collapse
Affiliation(s)
- Kanishka Rangamuwa
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine (RMH), University of Melbourne, Parkville, Australia
| | - Tracy Leong
- Department of Respiratory Medicine, Austin Hospital, Heidelberg, Victoria, Australia
| | - Clare Weeden
- Personalised Oncology Division, Walter Eliza Hall institute, Melbourne, Australia
| | | | - Steven Bozinovski
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Michael Christie
- Department of Pathology, Royal Melbourne Hospital, Melbourne, Australia
| | - Tom John
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Phillip Antippa
- Department of Thoracic Surgery, Royal Melbourne Hospital, Melbourne, Australia
| | - Louis Irving
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Australia
| | - Daniel Steinfort
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Australia.,Department of Medicine (RMH), University of Melbourne, Parkville, Australia
| |
Collapse
|
22
|
Xu Y, Song G, Xie S, Jiang W, Chen X, Chu M, Hu X, Wang ZW. The roles of PD-1/PD-L1 in the prognosis and immunotherapy of prostate cancer. Mol Ther 2021; 29:1958-1969. [PMID: 33932597 DOI: 10.1016/j.ymthe.2021.04.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple studies have confirmed that programmed cell death 1/programmed cell death ligand-1 (PD-1/PD-L1) and immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 play pivotal roles in the treatment of numerous tumors. Patients suffering from cancer are provided hope in the form of immunotherapy. In this review, we discuss the finding that high PD-L1 expression is associated with poor clinical outcomes in prostate cancer patients. Some molecules exert their antitumor effects by downregulating PD-L1 expression in prostate cancer. Additionally, we discuss and summarize the important roles played by anti-PD-1/PD-L1 immunotherapy and its combination with other drugs, including chemotherapy and vaccines, in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Yichi Xu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Gendi Song
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shangdan Xie
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xin Chen
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Man Chu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaoli Hu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| | - Zhi-Wei Wang
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China; Center of Scientific Research, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|
23
|
Oligometastatic Disease and Interventional Oncology: Rationale and Research Directions. ACTA ACUST UNITED AC 2021; 26:166-173. [PMID: 32205542 DOI: 10.1097/ppo.0000000000000442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oligometastatic disease (OMD) is generally defined as a stage of clinically or radiographically demonstrated metastatic disease limited in total disease burden and without rapid spread. Interventional oncology performs local therapies for primary and metastatic cancers, including OMD. Interventional oncology treatments can be pursued both as definitive therapy and for palliative purposes. Applied to OMD, these interventions can offer patients a decreasing overall tumor burden, minimizing cancer morbidity, and early evidence suggests a survival benefit. Here, we discuss the range of interventional oncology treatments, including ablation, chemoembolization, radioembolization, and irreversible electroporation. We describe the rationale for their application to OMD and discuss future directions for research.
Collapse
|
24
|
Connor MJ, Shah TT, Smigielska K, Day E, Sukumar J, Fiorentino F, Sarwar N, Gonzalez M, Falconer A, Klimowska-Nassar N, Evans M, Naismith OF, Thippu Jayaprakash K, Price D, Gayadeen S, Basak D, Horan G, McGrath J, Sheehan D, Kumar M, Ibrahim A, Brock C, Pearson RA, Anyamene N, Heath C, Shergill I, Rai B, Hellawell G, McCracken S, Khoubehi B, Mangar S, Khoo V, Dudderidge T, Staffurth JN, Winkler M, Ahmed HU. Additional Treatments to the Local tumour for metastatic prostate cancer-Assessment of Novel Treatment Algorithms (IP2-ATLANTA): protocol for a multicentre, phase II randomised controlled trial. BMJ Open 2021; 11:e042953. [PMID: 33632752 PMCID: PMC7908915 DOI: 10.1136/bmjopen-2020-042953] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/08/2021] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Survival in men diagnosed with de novo synchronous metastatic prostate cancer has increased following the use of upfront systemic treatment, using chemotherapy and other novel androgen receptor targeted agents, in addition to standard androgen deprivation therapy (ADT). Local cytoreductive and metastasis-directed interventions are hypothesised to confer additional survival benefit. In this setting, IP2-ATLANTA will explore progression-free survival (PFS) outcomes with the addition of sequential multimodal local and metastasis-directed treatments compared with standard care alone. METHODS A phase II, prospective, multicentre, three-arm randomised controlled trial incorporating an embedded feasibility pilot. All men with new histologically diagnosed, hormone-sensitive, metastatic prostate cancer, within 4 months of commencing ADT and of performance status 0 to 2 are eligible. Patients will be randomised to Control (standard of care (SOC)) OR Intervention 1 (minimally invasive ablative therapy to prostate±pelvic lymph node dissection (PLND)) OR Intervention 2 (cytoreductive radical prostatectomy±PLND OR prostate radiotherapy±pelvic lymph node radiotherapy (PLNRT)). Metastatic burden will be prespecified using the Chemohormonal Therapy Versus Androgen Ablation Randomized Trial for Extensive Disease (CHAARTED) definition. Men with low burden disease in intervention arms are eligible for metastasis-directed therapy, in the form of stereotactic ablative body radiotherapy (SABR) or surgery. Standard systemic therapy will be administered in all arms with ADT±upfront systemic chemotherapy or androgen receptor agents. Patients will be followed-up for a minimum of 2 years. PRIMARY OUTCOME PFS. Secondary outcomes include predictive factors for PFS and overall survival; urinary, sexual and rectal side effects. Embedded feasibility sample size is 80, with 918 patients required in the main phase II component. Study recruitment commenced in April 2019, with planned follow-up completed by April 2024. ETHICS AND DISSEMINATION Approved by the Health Research Authority (HRA) Research Ethics Committee Wales-5 (19/WA0005). Study results will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT03763253; ISCRTN58401737.
Collapse
Affiliation(s)
- Martin John Connor
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial Urology, Imperial College Healthcare NHS Trust, London, UK
| | - Taimur Tariq Shah
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Katarzyna Smigielska
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial College Clinical Trials Unit, Imperial College London, London, UK
| | - Emily Day
- Imperial College Clinical Trials Unit, Imperial College London, London, UK
| | - Johanna Sukumar
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial College Clinical Trials Unit, Imperial College London, London, UK
| | | | - Naveed Sarwar
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Michael Gonzalez
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Alison Falconer
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Natalia Klimowska-Nassar
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial College Clinical Trials Unit, Imperial College London, London, UK
| | - Martin Evans
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Olivia Frances Naismith
- Radiotherapy Trials Quality Assurance (RTTQA), Royal Marsden NHS Foundation Trust, London, UK
| | | | - Derek Price
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Shiva Gayadeen
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Dolan Basak
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Gail Horan
- Department of Oncology, Addenbrooke's Hospital, Cambridge, Cambridgeshire, UK
| | - John McGrath
- Department of Urology, Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, UK
| | - Denise Sheehan
- Department of Oncology, Royal Devon and Exeter NHS Foundation Trust, Exeter, Devon, UK
| | - Manal Kumar
- Department of Urology, Arrowe Park Hospital, Wirral University Teaching Hospital NHS Foundation Trust, Wirral, UK
| | - Azman Ibrahim
- Department of Clinical Oncology, Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, UK
| | - Cathryn Brock
- Department of Oncology, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Rachel A Pearson
- Department of Oncology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Nicola Anyamene
- Department of Oncology, London North West University Healthcare NHS Trust, Harrow, London, UK
| | - Catherine Heath
- Department of Radiotherapy, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Iqbal Shergill
- Department of Urology, Wrexham Maelor Hospital, Wrexham, UK
| | - Bhavan Rai
- Department of Urology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Giles Hellawell
- Department of Urology, Northwick Park Hospital, London North West University Healthcare NHS Trust, Harrow, London, UK
| | - Stuart McCracken
- Department of Urology, Sunderland Royal Hospital, Sunderland, UK
| | - Bijan Khoubehi
- Department of Urology, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Stephen Mangar
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Vincent Khoo
- Department of Oncology, The Royal Marsden NHS Foundation and Institute of Cancer Research, London, UK
| | - Tim Dudderidge
- Department of Urology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - John Nicholas Staffurth
- Research, Velindre Cancer Centre, Cardiff, UK
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Mathias Winkler
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial Urology, Imperial College Healthcare NHS Trust, London, UK
| | - Hashim Uddin Ahmed
- Imperial Prostate, Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Imperial Urology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
25
|
Yonezawa N, Murakami H, Demura S, Kato S, Miwa S, Yoshioka K, Shinmura K, Yokogawa N, Shimizu T, Oku N, Kitagawa R, Handa M, Annen R, Kurokawa Y, Fushimi K, Mizukoshi E, Tsuchiya H. Abscopal Effect of Frozen Autograft Reconstruction Combined with an Immune Checkpoint Inhibitor Analyzed Using a Metastatic Bone Tumor Model. Int J Mol Sci 2021; 22:1973. [PMID: 33671258 PMCID: PMC7922593 DOI: 10.3390/ijms22041973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/08/2021] [Accepted: 02/14/2021] [Indexed: 01/10/2023] Open
Abstract
We evaluated the abscopal effect of re-implantation of liquid nitrogen-treated tumor-bearing bone grafts and the synergistic effect of anti-PD-1 (programmed death-1) therapy using a bone metastasis model, created by injecting MMT-060562 cells into the bilateral tibiae of 6-8-week-old female C3H mice. After 2 weeks, the lateral tumors were treated by excision, cryotreatment using liquid nitrogen, excision with anti-PD-1 treatment, and cryotreatment with anti-PD-1 treatment. Anti-mouse PD-1 4H2 was injected on days 1, 6, 12, and 18 post-treatment. The mice were euthanized after 3 weeks; the abscopal effect was evaluated by focusing on growth inhibition of the abscopal tumor. The re-implantation of frozen autografts significantly inhibited the growth of the remaining abscopal tumors. However, a more potent abscopal effect was observed in the anti-PD-1 antibody group. The number of CD8+ T cells infiltrating the abscopal tumor and tumor-specific interferon-γ (IFN-γ)-producing spleen cells increased in the liquid nitrogen-treated group compared with those in the excision group, with no significant difference. The number was significantly higher in the anti-PD-1 antibody-treated group than in the non-treated group. Overall, re-implantation of tumor-bearing frozen autograft has an abscopal effect on abscopal tumor growth, although re-implantation of liquid nitrogen-treated bone grafts did not induce a strong T-cell response or tumor-suppressive effect.
Collapse
Affiliation(s)
- Noritaka Yonezawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Hideki Murakami
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan;
| | - Satoru Demura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Satoshi Kato
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Katsuhito Yoshioka
- Department of Orthopaedic Surgery, National Hospital Organization Kanazawa Medical Center, Kanazawa, Ishikawa 920-8650, Japan;
| | - Kazuya Shinmura
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Noriaki Yokogawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Takaki Shimizu
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Norihiro Oku
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Ryo Kitagawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Makoto Handa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Ryohei Annen
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Yuki Kurokawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| | - Kazumi Fushimi
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Ishikawa 920-8641, Japan; (K.F.); (E.M.)
| | - Eishiro Mizukoshi
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa, Ishikawa 920-8641, Japan; (K.F.); (E.M.)
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kanazawa University, 13-1 Takara-machi, Kanazawa 920-8641, Japan; (N.Y.); (S.K.); (S.M.); (K.S.); (N.Y.); (T.S.); (N.O.); (R.K.); (M.H.); (R.A.); (Y.K.); (H.T.)
| |
Collapse
|
26
|
wang N, Ye Y, Deng M, Zhao D, Jiang L, Chen D, Wu Z, Wang Y, Li Z, Yang Z, Li J, Zhou F, Li Y. Prostate cryoablation combined with androgen deprivation therapy for newly diagnosed metastatic prostate cancer: a propensity score-based study. Prostate Cancer Prostatic Dis 2021; 24:837-844. [PMID: 33664457 PMCID: PMC8384623 DOI: 10.1038/s41391-021-00335-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/22/2020] [Accepted: 01/27/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND Several studies showed that androgen deprivation therapy (ADT) plus local treatment of prostate could improve metastatic prostate cancer (mPCa) patients' survival. To date there are few studies analyzed the value of prostate cryoablation in mPCa. The objective of our analysis is to evaluate the oncological results and clinical value of prostate cryoablation combined with ADT compared with ADT alone in newly diagnosed mPCa patients. METHODS Newly diagnosed mPCa patients undergoing cryoablation plus ADT (group A) between January 2011 and November 2018 were identified. Patients receiving ADT alone (group B) were selected from the same institutional prostate cancer database by propensity score matching based on clinical characteristics. Oncological results and clinical value in symptom control and primary lesion treatment were compared. RESULTS Fifty-four patients were included in each group. Prostate cryoablation was well tolerated. The median follow-up time was 40 (27-53) and 39 (31-54) months in group A and group B, respectively. Patients in group A had a lower median prostate-specific antigen (PSA) nadir (0.025 ng/mL vs. 0.230 ng/mL, p = 0.001), longer median failure-free survival (FFS) (39 months vs. 21 months, p = 0.005), and median metastatic castration-resistant prostate cancer (mCRPC)-free survival (39 months vs. 21 months, p = 0.007). No difference in cancer-specific survival and overall survival was found between the two groups. Multivariate Cox analysis showed combination therapy reduced the risk of FFS by 45.8% (HR = 0.542 [95% CI 0.329-0.893]; p = 0.016). Patients in group A had better clinical relief of urinary symptoms (79.1 vs. 59.1%, p = 0.044) and required less treatment of primary lesions for symptomatic relief (13.0 vs. 31.5%, p = 0.021). CONCLUSIONS Prostate cryoablation plus ADT decreases PSA nadir, prolongs FFS and mCRPC-free survival, relieves urinary symptoms and reduces the need for treating primary lesions in newly diagnosed mPCa patients compared to ADT alone.
Collapse
Affiliation(s)
- Ning wang
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Yangtian Ye
- grid.412601.00000 0004 1760 3828Department of Urology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Minhua Deng
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Diwei Zhao
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Lijuan Jiang
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Dong Chen
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Zhiming Wu
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Yanjun Wang
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - ZhiYong Li
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Zhenyu Yang
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Jibin Li
- grid.488530.20000 0004 1803 6191Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Fangjian Zhou
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| | - Yonghong Li
- grid.488530.20000 0004 1803 6191Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China; Collaborative Innovation Cencer for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong China
| |
Collapse
|
27
|
Patel D, McKay R, Parsons JK. Immunotherapy for Localized Prostate Cancer: The Next Frontier? Urol Clin North Am 2020; 47:443-456. [PMID: 33008495 DOI: 10.1016/j.ucl.2020.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer vaccines, cytokines, and checkpoint inhibitors are immunotherapeutic agents that act within the cancer immunity cycle. Prostate cancer has provided unique opportunities for, and challenges to, immunotherapy drug development, including low tumor mutational burdens, limited expression of PD-L1, and minimal T-cell intratumoral infiltrates. Nevertheless, efforts are ongoing to help prime prostate tumors by turning a "cold" prostate cancer "hot" and thus rendering them more susceptible to immunotherapy. Combination treatments, use of molecular biomarkers, and use of new immunotherapeutic agents provide opportunities to enhance the immune response to prostate tumors.
Collapse
Affiliation(s)
- Devin Patel
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA
| | - Rana McKay
- Division of Hematology-Oncology, Department of Internal Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - J Kellogg Parsons
- Department of Urology, University of California San Diego, 9400 Campus Point Drive, MC7987, La Jolla, CA 92093, USA.
| |
Collapse
|
28
|
Qian L, Shen Y, Xie J, Meng Z. Immunomodulatory effects of ablation therapy on tumors: Potentials for combination with immunotherapy. Biochim Biophys Acta Rev Cancer 2020; 1874:188385. [PMID: 32554098 DOI: 10.1016/j.bbcan.2020.188385] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
|
29
|
Zhang X, Chen H, Li G, Zhou X, Shi Y, Zou F, Chen Y, Gao J, Yang S, Wu S, Long Z. Increased Tim-3 expression on TILs during treatment with the Anchored GM-CSF vaccine and anti-PD-1 antibodies is inversely correlated with response in prostate cancer. J Cancer 2020; 11:648-656. [PMID: 31942188 PMCID: PMC6959042 DOI: 10.7150/jca.29705] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
Programmed death receptor-1 (PD-1) and T cell immunoglobulin and mucin domain-containing protein-3 (Tim-3) play important roles in tumor immune evasion. PD-1 blockade could produce an effective antitumor effect in many solid tumors except prostate cancer (PCa) because of rare programmed death ligand-1 (PD-L1) expression on PCa cells. Streptavidin (SA)-GM-CSF surface-anchored tumor cell (Anchored GM-CSF) vaccines could increase the number of tumor-infiltrating lymphocytes (TILs) and induce specific antitumor immune responses. The Anchored-GM-CSF vaccine and anti-PD-1 antibodies exerted synergistic effects in mouse models of PCa metastasis. However, the response rate was low due to the presence of other negative regulatory pathways. Tim-3 expression could be upregulated at resistance to combination therapy with anti-PD-1 antibodies and the Anchored GM-CSF vaccine. Sequential administration of anti-PD-1 and anti-Tim-3 antibodies could further improve the efficacy of the Anchored GM-CSF vaccine therapy, and tumor regression was noted in over 60% of animals. This triple therapy improved the specific cytotoxic activity, proliferation and secretion of CD8+ TILs and reduced the production of tumor-promoting cytokines. These findings indicated that this triple therapy could induce a robust antitumor immune response in mouse models of PCa.
Collapse
Affiliation(s)
- Xinji Zhang
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Haixiong Chen
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Guanfeng Li
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Xiangyun Zhou
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Yuqiang Shi
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Feng Zou
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Yuanxiang Chen
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shaomin Yang
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
- ✉ Corresponding authors: Zhaolin Long and Shihao Wu, Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318701; fax: +86 0757-22318702; Email addresses: and ; Shaomin Yang, Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318611; fax: +86 0757-22318612; Email addresses:
| | - Shihao Wu
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
- ✉ Corresponding authors: Zhaolin Long and Shihao Wu, Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318701; fax: +86 0757-22318702; Email addresses: and ; Shaomin Yang, Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318611; fax: +86 0757-22318612; Email addresses:
| | - Zhaolin Long
- Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China
- ✉ Corresponding authors: Zhaolin Long and Shihao Wu, Department of Urology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318701; fax: +86 0757-22318702; Email addresses: and ; Shaomin Yang, Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde District, Foshan), Foshan, 528300, China; Phone: +86 0757-22318611; fax: +86 0757-22318612; Email addresses:
| |
Collapse
|
30
|
Ross AE, Hurley PJ, Tran PT, Rowe SP, Benzon B, Neal TO, Chapman C, Harb R, Milman Y, Trock BJ, Drake CG, Antonarakis ES. A pilot trial of pembrolizumab plus prostatic cryotherapy for men with newly diagnosed oligometastatic hormone-sensitive prostate cancer. Prostate Cancer Prostatic Dis 2019; 23:184-193. [PMID: 31611635 PMCID: PMC7031012 DOI: 10.1038/s41391-019-0176-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Monotherapy with immune checkpoint inhibitors has generally been unsuccessful in men with advanced prostate cancer. Preclinical data support the notion that cryotherapy may improve immune-mediated and anti-tumor responses. The objective of this study was to assess the safety and feasibility of whole-prostate gland cryotherapy combined with pembrolizumab and androgen deprivation in men with oligometastatic hormone-sensitive prostate cancer. METHODS This single-institution, pilot trial recruited 12 patients with newly diagnosed oligometastatic prostate cancer between 2015 and 2016. Patients underwent whole-prostate cryoablation combined with short-term androgen deprivation (eight months) and pembrolizumab (6 doses). The primary clinical endpoints were the number of patients with a PSA level of <0.6 ng/mL at one year and the frequency of adverse events. Other outcome measures included progression-free survival and systemic therapy-free survival. Exploratory analyses included PD-L1 protein expression. RESULTS Forty two percent (5/12) of patients had a PSAs of <0.6 ng/mL at one year though only 2 of these patients had recovered their testosterone at this time point. Median progression-free survival was 14 months, and median systemic therapy-free survival was 17.5 months. PD-L1 expression was not detectable by IHC in patients with evaluable tissue. All adverse events were grade ≤2, and there were no apparent complications from cryotherapy. CONCLUSIONS Whole-prostate cryoablation combined with short-term androgen deprivation and pembrolizumab treatment was well tolerated and no safety concerns were observed in men with oligometastatic prostate cancer. Though local disease appeared effectively treated in the majority of men, the regimen only infrequency led to sustained disease control following testosterone recovery.
Collapse
Affiliation(s)
- Ashley E Ross
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Paula J Hurley
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Phuoc T Tran
- The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Department of Radiation Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Benjamin Benzon
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tanya O' Neal
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Carolyn Chapman
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rana Harb
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yelena Milman
- The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Bruce J Trock
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Charles G Drake
- The Department of Medicine, Columbia University, New York, NY, USA
| | - Emmanuel S Antonarakis
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.,The Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
31
|
Yu X, Liang C, Zhang Y, Zhang W, Chen H. Inhibitory short peptides targeting EPS8/ABI1/SOS1 tri-complex suppress invasion and metastasis of ovarian cancer cells. BMC Cancer 2019; 19:878. [PMID: 31488087 PMCID: PMC6727365 DOI: 10.1186/s12885-019-6087-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/23/2019] [Indexed: 01/13/2023] Open
Abstract
Background We aimed to develop inhibitory short peptides that can prevent protein interactions of SOS1/EPS8/ABI1 tri-complex, a key component essential for ovarian cancer metastasis. Methods Plasmids containing various regions of HA-tagged ABI1 were co-transfected into ovarian cancer cells with Flag-tagged SOS1 or Myc-tagged EPS8. Co-immunoprecipitation and GST-pulldown assay were used to identify the regions of ABI1 responsible for SOS1 and EPS8 binding. Inhibitory short peptides of these binding regions were synthesized and modified with HIV-TAT sequence. The blocking effects of the peptides on ABI1-SOS1 or ABI1-EPS8 interactions in vitro and in vivo were determined by GST-pulldown assay. The capability of these short peptides in inhibiting invasion and metastasis of ovarian cancer cell was tested by Matrigel invasion assay and peritoneal metastatic colonization assay. Results The formation of endogenous SOS1/EPS8/ABI1 tri-complex was detected in the event of LPA-induced ovarian cancer cell invasion. In the tri-complex, ABI1 acted as a scaffold protein holding together SOS1 and EPS8. The SH3 and poly-proline+PxxDY regions of ABI1 were responsible for SOS1 and EPS8 binding, respectively. Inhibitory short peptides p + p-8 (ppppppppvdyedee) and SH3–3 (ekvvaiydytkdkddelsfmegaii) could block ABI1-SOS1 and ABI1-EPS8 interaction in vitro. TAT-p + p-8 peptide could disrupt ABI1-EPS8 interaction and suppress the invasion and metastasis of ovarian cancer cells in vivo. Conclusions TAT-p + p-8 peptide could efficiently disrupt the ABI1-EPS8 interaction, tri-complex formation, and block the invasion and metastasis of ovarian cancer cells.
Collapse
Affiliation(s)
- Xuechen Yu
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Chuan Liang
- Department of Cardiothoracic vascular surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Yuanzhen Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Wei Zhang
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
| | - Huijun Chen
- Department of Gynaecology and Obstetrics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
32
|
Cryo combo augments ICI. Nat Rev Urol 2019; 15:336. [PMID: 29674670 DOI: 10.1038/s41585-018-0009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
33
|
T cells CD4+/CD8+ local immune modulation by prostate cancer hemi-cryoablation. World J Urol 2019; 38:673-680. [PMID: 31263944 DOI: 10.1007/s00345-019-02861-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/24/2019] [Indexed: 12/30/2022] Open
Abstract
PURPOSE Tumors escape from the immune system by decreasing CD8+ and increasing CD4+ T cells' activity, druggable targets. Thermal ablation might activate tumor-specific T cells by raising the presentation of tumor-specific antigens and hindering tumor negative immune regulation. Our aim was to assess T cell infiltrate pre- and post-cryoablation in a prospective observational study. METHODS A total of 240 sextant prostate biopsies cores (12 cores/patient) were collected from 10 unilateral prostate cancer patients (T1c, PSA density < 0.15 ng/dL, Gleason grade group 1, ≤ 2 cancer biopsy cores, and < 50% cancer core involvement) at diagnosis and 12 months after hemi-cryoablation. Cancer-positive (Diag+) and cancer-negative (Diag-) lobes at diagnosis and the same areas 12 months after hemi-cryoablation (Cryo+ and Cryo-, respectively) were explored by immunohistochemistry for infiltrating CD4+ and CD8+ T cells (in 45 random fields per prostate lobe, 400× magnification). The quantitative analysis of cells/mm2 and CD4+/CD8+ ratio were performed and compared among Diag+, Diag-, Cryo+, and Cryo- using ImageJ software. RESULTS There was a significant increase in tumor-infiltrating CD8+ T cells/mm2 in the Cryo+ tissue (mean, SD 0.31, 0.30) compared to Diag+ (0.18, 0.15), p = 0.015; confirmed in prostate acini (hot spots), p = 0.029, in which infiltrating CD4+/CD8+ T cells' ratio decreased after hemi-cryoablation, p = 0.006. Infiltrating CD4+ T cells/mm2 presented a trend to decrease in Cryo+ (0.26, 0.27) compared to Diag+ (0.38, 0.32). CONCLUSIONS This is the first study to show local immune modulation after prostate cancer cryoablation, characterized by decreasing CD4+/CD8+ T cells' ratio, potential for clinical impact by unleashing the T-cell response to cancer. Future studies are necessary to explore different energies and longer follow-up clinical endpoints.
Collapse
|
34
|
Aarts BM, Klompenhouwer EG, Rice SL, Imani F, Baetens T, Bex A, Horenblas S, Kok M, Haanen JBAG, Beets-Tan RGH, Gómez FM. Cryoablation and immunotherapy: an overview of evidence on its synergy. Insights Imaging 2019; 10:53. [PMID: 31111237 PMCID: PMC6527672 DOI: 10.1186/s13244-019-0727-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/25/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer cells can escape the immune system by different mechanisms. The evasion of cancer cells from immune surveillance is prevented by immune checkpoint inhibitors, allowing the patient’s own immune system to attack their cancer. Immune checkpoint inhibitors have shown improvement in overall survival for melanoma, lung cancer and renal cell carcinoma in clinical trials. Unfortunately, not all patients respond to this therapy. In cancer management, percutaneous ablation techniques are well established for both cure and local control of many tumour types. Cryoablation of the tumour tissue results in cell destruction by freezing. Contrary to heat-based ablative modalities, cryoablation induces tumour cell death by osmosis and necrosis. It is hypothesised that with necrosis, the intracellular contents of the cancer cells stay intact allowing the immune system to induce an immune-specific reaction. This immune-specific reaction can, in theory, also affect cancer cells outside the ablated tissue, known as the abscopal effect. Unfortunately, this effect is rarely observed, but when cryoablation is combined with immunotherapy, the effect of both therapies may be enhanced. Although several preclinical studies demonstrated a synergistic effect between cryoablation and immunotherapy, prospective clinical trials are needed to prove this clinical benefit for patients. In this review, we will outline the current evidence for the combination of cryoablation with immunotherapy to treat cancer.
Collapse
Affiliation(s)
- B M Aarts
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands. .,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
| | - E G Klompenhouwer
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - S L Rice
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Department of Radiology, Interventional Radiology Service, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, USA
| | - F Imani
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - T Baetens
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - A Bex
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - S Horenblas
- Department of Urology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - M Kok
- Department of Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - J B A G Haanen
- Department of Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - R G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
| | - F M Gómez
- Department of Radiology, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Department of Interventional Radiology, Hospital Clinic Universitari, Carrer de Villarroel 170, 08036, Barcelona, Spain
| |
Collapse
|