1
|
IJff M, Mei X, Scutigliani EM, Rodermond HM, van Bochove GGW, Krawczyk PM, Franken NAP, Stalpers LJA, Crezee J, Oei AL. Addition of PARP1-inhibition enhances chemoradiotherapy and thermoradiotherapy when treating cervical cancer in an in vivo mouse model. Int J Hyperthermia 2025; 42:2450514. [PMID: 39837264 DOI: 10.1080/02656736.2025.2450514] [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: 02/01/2024] [Revised: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Background: Efficacy of current treatment options for cervical cancer require improvement. Previous in vitro studies have shown the enhancing effects of the addition of PARP1-inhibitors to chemoradiotherapy and thermoradiotherapy. The aim of our present study was to test efficacy of different combinations of treatment modalities radiotherapy, cisplatin, hyperthermia and PARP1-inhibitors using in vitro tumor models, ex vivo treated patient samples and in vivo tumor models. Materials and Methods: In vitro clonogenic survival curves (0-6 Gy) show that PARP1-i (4-5 M Olaparib) enhances both chemoradiotherapy (0.3-0.5 µM cisplatin) and thermoradiotherapy (42 °C for 1 h) in SiHa, CaSki and HeLa cells. A cervical cancer mouse model and freshly obtained in-house developed patient-derived organoids were used to examine the effects of different treatment combinations. For the in vivo study, human cervical cancer (SiHa) cells were injected in the right hind leg of athymic nude mice. In vivo mouse experiments show that PARP1-i enhances thermoradiotherapy or chemoradiotherapy by reduction of tumor volumes. Five cycles of treatment were applied with the following doses per cycle: irradiation 3 Gy, hyperthermia 1 h at 42 °C, cisplatin at 2 mg/kg, and twice PARP1-i at 50 mg/kg. Results: Quadruple treatment, combining radiotherapy, hyperthermia, cisplatin and PARP1-i, was very effective but also lead to severe side effects causing severe weight loss and death. In contrast, thermoradiotherapy or chemoradiotherapy with addition of PARP1-i, were effective without serious side effects. Conclusion: The triple combinations are promising options for potentially more effective treatment of locally advanced cervical cancer without more toxicity.
Collapse
Affiliation(s)
- Marloes IJff
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Xionge Mei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Enzo M Scutigliani
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans M Rodermond
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Gregor G W van Bochove
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Przemek M Krawczyk
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas A P Franken
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Lukas J A Stalpers
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| | - Arlene L Oei
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiation Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Valable S, Césaire M, Lecrosnier K, Gilbert A, Tudor M, Vares G, Hamdi DH, Diouf OB, Nguyen Pham T, Coupey J, Thariat J, Lesueur P, Pérès EA, Aury-Landas J, Nikitaki Z, Haghdoost S, Laurent C, Poully JC, Balosso J, Bernaudin M, Savu DI, Chevalier F. Particle Therapy to Overcome Cancer Radiation Resistance: "ARCHADE" Consortium Updates in Radiation Biology. Cancers (Basel) 2025; 17:1580. [PMID: 40361506 PMCID: PMC12071746 DOI: 10.3390/cancers17091580] [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] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Radiation therapy is a medical treatment that uses high doses of radiation to kill or damage cancer cells. It works by damaging the DNA within the cancer cells, ultimately causing cell death. Radiotherapy can be used as a primary treatment, adjuvant treatment in combination with surgery or chemotherapy or palliative treatment to relieve symptoms in advanced cancer stages. Radiation therapy is constantly improving in order to enhance the effect on cancer cells and reduce the side effects on healthy tissues. Our results clearly demonstrate that proton therapy and, even more, carbon ion therapy appear as promising alternatives to overcome the radioresistance of various tumors thanks to less dependency on oxygen and a better ability to kill cancer stem cells. Interestingly, hadrons also retain the advantages of radiosensitization approaches. These data confirm the great ability of hadrons to spare healthy tissue near the tumor via various mechanisms (reduced lymphopenia, bystander effect, etc.). Technology and machine improvements such as image-guided radiotherapy or particle therapies can improve treatment quality and efficacy (dose deposition and biological effect) in tumors while increasingly sparing healthy tissues. Radiation biology can help to understand how cancer cells resist radiation (hypoxia, DNA repair mechanisms, stem cell status, cell cycle position, etc.), how normal tissues may display sensitivity to radiation and how radiation effects can be increased with either radiosensitizers or accelerated particles. All these research topics are under investigation within the ARCHADE research community in France. By focusing on these areas, radiotherapy can become more effective, targeted and safe, enhancing the overall treatment experience and outcomes for cancer patients. Our goal is to provide biological evidence of the therapeutic advantages of hadrontherapy, according to the tumor characteristics. This article aims to give an updated view of our research in radiation biology within the frame of the French "ARCHADE association" and new perspectives on research and treatment with the C400 multi-ions accelerator prototype.
Collapse
Affiliation(s)
- Samuel Valable
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Mathieu Césaire
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Kilian Lecrosnier
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Antoine Gilbert
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania;
- Faculty of Biology, University of Bucharest, 030018 Bucharest, Romania
| | - Guillaume Vares
- Autorité de Sûreté Nucléaire et de Radioprotection (ASNR), PSE-SANTE/SESANE/LRTOX, 92260 Fontenay-aux-Roses, France;
| | - Dounia Houria Hamdi
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Ousseynou Ben Diouf
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal
| | - Thao Nguyen Pham
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Julie Coupey
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Juliette Thariat
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre François Baclesse, 14000 Caen, France
| | - Paul Lesueur
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre Guillaume Le Conquérant, 76600 Le Havre, France
| | - Elodie Anne Pérès
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Juliette Aury-Landas
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Zacharenia Nikitaki
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Siamak Haghdoost
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Carine Laurent
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Normandie University, UNICAEN, UNIROUEN, ABTE UR4651, Cancer Center François Baclesse, 14000 Caen, France
| | - Jean-Christophe Poully
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| | - Jacques Balosso
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Radiation Oncology Department, Centre François Baclesse, 14000 Caen, France
| | - Myriam Bernaudin
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, 14000 Caen, France
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania;
| | - François Chevalier
- ARCHADE, Association for “Advanced Resource Center for Hadrontherapy in Europe”, 14000 Caen, France; (S.V.); (M.C.); (K.L.); (A.G.); (J.T.); (P.L.); (E.A.P.); (J.A.-L.); (Z.N.); (S.H.); (C.L.); (J.-C.P.); (J.B.); (M.B.)
- Université de Caen Normandie, ENSICAEN, CNRS, CEA, Normandie Université, CIMAP UMR6252, 14000 Caen, France; (D.H.H.); (O.B.D.)
| |
Collapse
|
3
|
Lei XY, He KY, Li QT, Zhang L, Wu DH, Yang JY, Guo JR, Liu MJ, Zhao ZL, Li JQ, Liu H, Zhao Y, Li YJ, Sun QH, Wu CG, Wang YF, Cao GS, Wang G, Jian YP, Xu ZX. PARylation of HMGA1 desensitizes esophageal squamous cell carcinoma to olaparib. Clin Transl Med 2024; 14:e70111. [PMID: 39690136 DOI: 10.1002/ctm2.70111] [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: 04/11/2024] [Revised: 11/01/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
As a chromatin remodelling factor, high mobility group A1 (HMGA1) plays various roles in both physiological and pathological conditions. However, its role in DNA damage response and DNA damage-based chemotherapy remains largely unexplored. In this study, we report the poly ADP-ribosylation (PARylation) of HMGA1 during DNA damage, leading to desensitization of esophageal squamous cell carcinoma (ESCC) cells to the poly(ADP-ribose) polymerase 1 (PARP1) inhibitor, olaparib. We found that HMGA1 accumulates at sites of DNA damage, where it interacts with PARP1 and undergoes PARylation at residues E47 and E50 in its conserved AT-hook domain. This modification enhances the accumulation of Ku70/Ku80 at the site of DNA damage and activates the DNA-dependent protein kinase catalytic subunit, facilitating nonhomologous end-joining repair. In both subcutaneous tumour models and genetically engineered mouse models of in situ esophageal cancer, HMGA1 interference increased tumour sensitivity to olaparib. Moreover, HMGA1 was highly expressed in ESCC tissues and positively correlated with PARP1 levels as well as poor prognosis in ESCC patients. Taken together, these findings reveal a mechanistic link between HMGA1 and PARP1 in regulating cell responses to DNA damage and suggest that targeting HMGA1 could be a promising strategy to increase cancer cell sensitivity to olaparib.
Collapse
Affiliation(s)
- Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, China
| | - Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jing-Yu Yang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jin-Rong Guo
- School of Life Sciences, Henan University, Kaifeng, China
| | - Meng-Jie Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yuan Zhao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yu-Jia Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, China
| | - Chen-Guang Wu
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yun-Fan Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Geng-Sheng Cao
- School of Life Sciences, Henan University, Kaifeng, China
| | - Gang Wang
- School of Life Sciences, Henan University, Kaifeng, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, China
| |
Collapse
|
4
|
Rødland GE, Temelie M, Eek Mariampillai A, Hauge S, Gilbert A, Chevalier F, Savu DI, Syljuåsen RG. Potential Benefits of Combining Proton or Carbon Ion Therapy with DNA Damage Repair Inhibitors. Cells 2024; 13:1058. [PMID: 38920686 PMCID: PMC11201490 DOI: 10.3390/cells13121058] [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: 04/29/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The use of charged particle radiotherapy is currently increasing, but combination therapy with DNA repair inhibitors remains to be exploited in the clinic. The high-linear energy transfer (LET) radiation delivered by charged particles causes clustered DNA damage, which is particularly effective in destroying cancer cells. Whether the DNA damage response to this type of damage is different from that elicited in response to low-LET radiation, and if and how it can be targeted to increase treatment efficacy, is not fully understood. Although several preclinical studies have reported radiosensitizing effects when proton or carbon ion irradiation is combined with inhibitors of, e.g., PARP, ATR, ATM, or DNA-PKcs, further exploration is required to determine the most effective treatments. Here, we examine what is known about repair pathway choice in response to high- versus low-LET irradiation, and we discuss the effects of inhibitors of these pathways when combined with protons and carbon ions. Additionally, we explore the potential effects of DNA repair inhibitors on antitumor immune signaling upon proton and carbon ion irradiation. Due to the reduced effect on healthy tissue and better immune preservation, particle therapy may be particularly well suited for combination with DNA repair inhibitors.
Collapse
Affiliation(s)
- Gro Elise Rødland
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Mihaela Temelie
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Adrian Eek Mariampillai
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France (F.C.)
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France (F.C.)
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
5
|
Kerkhove L, Geirnaert F, Dufait I, De Ridder M. Ferroptosis: Frenemy of Radiotherapy. Int J Mol Sci 2024; 25:3641. [PMID: 38612455 PMCID: PMC11011408 DOI: 10.3390/ijms25073641] [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: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
Recently, it was established that ferroptosis, a type of iron-dependent regulated cell death, plays a prominent role in radiotherapy-triggered cell death. Accordingly, ferroptosis inducers attracted a lot of interest as potential radio-synergizing drugs, ultimately enhancing radioresponses and patient outcomes. Nevertheless, the tumor microenvironment seems to have a major impact on ferroptosis induction. The influence of hypoxic conditions is an area of interest, as it remains the principal hurdle in the field of radiotherapy. In this review, we focus on the implications of hypoxic conditions on ferroptosis, contemplating the plausibility of using ferroptosis inducers as clinical radiosensitizers. Furthermore, we dive into the prospects of drug repurposing in the domain of ferroptosis inducers and radiosensitizers. Lastly, the potential adverse effects of ferroptosis inducers on normal tissue were discussed in detail. This review will provide an important framework for subsequent ferroptosis research, ascertaining the feasibility of ferroptosis inducers as clinical radiosensitizers.
Collapse
Affiliation(s)
| | | | | | - Mark De Ridder
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (L.K.); (F.G.); (I.D.)
| |
Collapse
|
6
|
Park S, Choi C, Kim H, Shin YJ, Oh Y, Park W, Cho WK, Kim N. Olaparib enhances sensitization of BRCA-proficient breast cancer cells to x-rays and protons. Breast Cancer Res Treat 2024; 203:449-461. [PMID: 37902934 DOI: 10.1007/s10549-023-07150-4] [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: 06/08/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023]
Abstract
PURPOSE This study aimed to compare the radiosensitizing effect of the PARP inhibitor, Olaparib, between proton and X-rays irradiations in BRCA-proficient breast cancer (BC) cells. METHODS Two BRCA-proficient BC cell lines, MDA-MB-231 and T47D BC, were used. Cell proliferation was assessed using the CCK-8 assay, and radiosensitivity was determined through the clonogenic survival assay. Flow cytometry was employed to analyze cell cycle distribution and apoptosis. The kinetics of DNA damage repair were evaluated using γH2AX immunofluorescence imaging and the comet assay. Tumor spheroid assays were conducted to test radiosensitivity in a three-dimensional culture condition. RESULTS Olaparib sensitized both MDA-MB-231 and T47D cells to proton and X-ray irradiation in the clonogenic assay. MDA-MB-231 cells exhibited a higher dose enhancement factor for Olaparib than T47D cells. Olaparib increased radiation-induced G2/M cell cycle arrest and apoptosis specifically in MDA-MB-231 cells. γH2AX immunostaining and the comet assay showed Olaparib augmented radiation-induced DNA damage and apoptosis. The enhancement effect of Olaparib was more pronounced in proton irradiation than in X-ray irradiation, particularly in MDA-MB-231 cells than T47D cells. Both radiation and Olaparib dose-dependently inhibited spheroid growth in both cell lines. The synergy scores demonstrated that Olaparib interacted more strongly with protons than X-rays. The addition of an ATR inhibitor further enhanced Olaparib-induced proton radiosensitization in MDA-MB-231 cells. CONCLUSION This study found that Olaparib enhanced radiation efficacy in BRCA-proficient breast cancer cells, with a more pronounced effect observed with proton irradiation compared to X-ray irradiation. Combining Olaparib with an ATR inhibitor increased the radiosensitizing effect of protons.
Collapse
Affiliation(s)
- Sohee Park
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Changhoon Choi
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| | - Haeyoung Kim
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Yong Jae Shin
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Yunjeong Oh
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Won Park
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
- Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Won Kyung Cho
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Nalee Kim
- Department of Radiation Oncology, Samsung Medical Center, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| |
Collapse
|
7
|
Brand NR, Yang YW, Ding V, Dutta H, Peto CJ, Lemjabbar-Alaoui H, Jablons DM. Novel dual action PARP and microtubule polymerization inhibitor AMXI-5001 powerfully inhibits growth of esophageal carcinoma both alone and in combination with radiotherapy. Am J Cancer Res 2024; 14:378-389. [PMID: 38323288 PMCID: PMC10839305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/27/2023] [Indexed: 02/08/2024] Open
Abstract
Esophageal cancer is one of the leading causes of cancer deaths globally with an incidence that is concentrated in specific hot spots in Eastern Asia, the Middle East, Eastern Africa, and South America. 10-year overall survival for patients treated with standard of care chemoradiation followed by surgical resection is below 40% highlighting the need for novel therapeutics to treat this disease. We assessed the effect of AMXI-5001, a novel small molecule poly ADP-Ribose polymerase (PARP) inhibitor and microtubule polymerization inhibitor on tumor growth inhibition in both in-vitro and in-vivo murine models. We found that AMXI-5001 was the most potent growth inhibitor of 8 out of 9 different esophageal carcinoma cell lines compared to other clinically available PARP inhibitors, Olaparib, Niraparib, Rucaparib, and Talazoparib. We then confirmed the previously described mechanism of action of AMXI-5001 as a PARP-inhibitor and microtubule polymerization inhibitor using both a PARP trapping assay and immunofluorescence. To further assess AMXI-5001's potential as a therapeutic for esophageal carcinoma we evaluated the effect of AMXI-5001 in combination with standard chemotherapy agents, Cisplatin and 5 Fluorouracil. We showed that AMXI-5001 synergistically inhibits growth in KYSE-70, a squamous esophageal cell line in combination with these drugs. In addition, we found that AMXI-5001 was an effective radiosensitizer, and squamous esophageal carcinoma cell lines treated 24 hours prior to external beam radiation showed significantly more growth inhibition compared to controls. Finally, we assessed the effect of AMXI-5001 monotherapy and in combination with radiotherapy in a xenograft mouse model implanted with subcutaneous KYSE-70 cells. Compared to vehicle control, and those treated with either AMXI-5001 alone or radiation alone, mice treated with both AMXI-5001 and radiation had significant tumor response. In conclusion, AMXI-5001 is an orally bioavailable dual-action PARP and microtubule polymerization inhibitor that holds promise in the treatment of esophageal carcinoma.
Collapse
Affiliation(s)
- Nathan R Brand
- Department of Surgery, University of CaliforniaSan Francisco, California, USA
| | - Yi-Wei Yang
- Thoracic Oncology Laboratory, University of CaliforniaSan Francisco, California, USA
| | - Vivianne Ding
- Thoracic Oncology Laboratory, University of CaliforniaSan Francisco, California, USA
| | - Hannah Dutta
- Thoracic Oncology Laboratory, University of CaliforniaSan Francisco, California, USA
| | - Csaba J Peto
- Thoracic Oncology Laboratory, University of CaliforniaSan Francisco, California, USA
| | | | - David M Jablons
- Department of Surgery, University of CaliforniaSan Francisco, California, USA
| |
Collapse
|
8
|
Sun C, Chu A, Song R, Liu S, Chai T, Wang X, Liu Z. PARP inhibitors combined with radiotherapy: are we ready? Front Pharmacol 2023; 14:1234973. [PMID: 37954854 PMCID: PMC10637512 DOI: 10.3389/fphar.2023.1234973] [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: 06/05/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
PARP was an enzyme found in the nucleus of eukaryotic cells that played a crucial role in repairing damaged DNA. Recently, PARP inhibitors have demonstrated great potential in cancer treatment. Thus, the FDA has approved several small-molecule PARP inhibitors for cancer maintenance therapy. The combination of PARP inhibitors and radiotherapy relies on synthetic lethality, taking advantage of the flaws in DNA repair pathways to target cancer cells specifically. Studies conducted prior to clinical trials have suggested that the combination of PARP inhibitors and radiotherapy can enhance the sensitivity of cancer cells to radiation, intensify DNA damage, and trigger cell death. Combining radiotherapy with PARP inhibitors in clinical trials has enhanced the response rate and progression-free survival of diverse cancer patients. The theoretical foundation of PARP inhibitors combined with radiotherapy is explained in detail in this article, and the latest advances in preclinical and clinical research on these inhibitors for tumor radiotherapy are summarized. The problems in the current field are recognized in our research and potential therapeutic applications for tumors are suggested. Nevertheless, certain obstacles need to be tackled when implementing PARP inhibitors and radiotherapies in clinical settings. Factors to consider when using the combination therapy are the most suitable schedule and amount of medication, identifying advantageous candidates, and the probable adverse effects linked with the combination. The combination of radiotherapy and PARP inhibitors can greatly enhance the effectiveness of cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Xin Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zongwen Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Kheyrandish MR, Mir SM, Sheikh Arabi M. DNA repair pathways as a novel therapeutic strategy in esophageal cancer: A review study. Cancer Rep (Hoboken) 2022; 5:e1716. [PMID: 36147024 PMCID: PMC9675361 DOI: 10.1002/cnr2.1716] [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: 11/17/2021] [Revised: 08/02/2022] [Accepted: 09/07/2022] [Indexed: 01/25/2023] Open
Abstract
Esophageal cancer (EC) is a common malignancy with a poor prognosis worldwide. There are two core pathways that repair double-strand breaks, homologous recombination (HR) and non-homologous end joining (NHEJ) and numerous proteins are recognized that affect the occurrence of HR and NHEJ. Altered DNA damage response (DDR) pathways are associated with cancer susceptibility and affect therapeutic response and resistance in cancers. DDR pathway alterations in EC are still poorly understood. Therefore, the identification of alterations in specific genes in DDR pathways may potentially result in novel treatments for resistant cancers, especially EC. In this review, we aimed to focus on different aspects of DNA damage and repair processes in EC. Also, we reviewed new therapeutic strategies via targeting DNA repair machinery components.
Collapse
Affiliation(s)
| | - Seyed Mostafa Mir
- Metabolic Disorders Research CenterGolestan University of Medical SciencesGorganIran,Department of Clinical Biochemistry, Faculty of MedicineGolestan University of Medical SciencesGorganIran
| | - Mehdi Sheikh Arabi
- Medical Cellular and Molecular Research CenterGolestan University of Medical SciencesGorganIran
| |
Collapse
|
10
|
Kowalchuk RO, Corbin KS, Jimenez RB. Particle Therapy for Breast Cancer. Cancers (Basel) 2022; 14:cancers14041066. [PMID: 35205814 PMCID: PMC8870138 DOI: 10.3390/cancers14041066] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
Particle therapy has received increasing attention in the treatment of breast cancer due to its unique physical properties that may enhance patient quality of life and reduce the late effects of therapy. In this review, we will examine the rationale for the use of proton and carbon therapy in the treatment of breast cancer and highlight their potential for sparing normal tissue injury. We will discuss the early dosimetric and clinical studies that have been pursued to date in this domain before focusing on the remaining open questions limiting the widespread adoption of particle therapy.
Collapse
Affiliation(s)
- Roman O. Kowalchuk
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (R.O.K.); (K.S.C.)
| | - Kimberly S. Corbin
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA; (R.O.K.); (K.S.C.)
| | - Rachel B. Jimenez
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA 02114, USA
- Correspondence:
| |
Collapse
|
11
|
Principe DR. Precision Medicine for BRCA/PALB2-Mutated Pancreatic Cancer and Emerging Strategies to Improve Therapeutic Responses to PARP Inhibition. Cancers (Basel) 2022; 14:cancers14040897. [PMID: 35205643 PMCID: PMC8869830 DOI: 10.3390/cancers14040897] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary For the small subset of pancreatic ductal adenocarcinoma (PDAC) patients with loss-of-function mutations to BRCA1/2 or PALB2, both first-line and maintenance therapy differs significantly. These mutations confer a loss of double-strand break DNA homologous recombination (HR), substantially altering drug sensitivities. In this review, we discuss the current treatment guidelines for PDAC tumors deficient in HR, as well as newly emerging strategies to improve drug responses in this population. We also highlight additional patient populations in which these strategies may also be effective, and novel strategies aiming to confer similar drug sensitivity to tumors proficient in HR repair. Abstract Pancreatic cancer is projected to become the second leading cause of cancer-related death by 2030. As patients typically present with advanced disease and show poor responses to broad-spectrum chemotherapy, overall survival remains a dismal 10%. This underscores an urgent clinical need to identify new therapeutic approaches for PDAC patients. Precision medicine is now the standard of care for several difficult-to-treat cancer histologies. Such approaches involve the identification of a clinically actionable molecular feature, which is matched to an appropriate targeted therapy. Selective poly (ADP-ribose) polymerase (PARP) inhibitors such as Niraparib, Olaparib, Talazoparib, Rucaparib, and Veliparib are now approved for several cancers with loss of high-fidelity double-strand break homologous recombination (HR), namely those with deleterious mutations to BRCA1/2, PALB2, and other functionally related genes. Recent evidence suggests that the presence of such mutations in pancreatic ductal adenocarcinoma (PDAC), the most common and lethal pancreatic cancer histotype, significantly alters drug responses both with respect to first-line chemotherapy and maintenance therapy. In this review, we discuss the current treatment paradigm for PDAC tumors with confirmed deficits in double-strand break HR, as well as emerging strategies to both improve responses to PARP inhibition in HR-deficient PDAC and confer sensitivity to tumors proficient in HR repair.
Collapse
Affiliation(s)
- Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
12
|
High-Throughput 3D Tumor Spheroid Array Platform for Evaluating Sensitivity of Proton-Drug Combinations. Int J Mol Sci 2022; 23:ijms23020587. [PMID: 35054773 PMCID: PMC8775525 DOI: 10.3390/ijms23020587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/25/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022] Open
Abstract
Proton beam therapy (PBT) is a critical treatment modality for head and neck squamous cell carcinoma (HNSCC). However, not much is known about drug combinations that may improve the efficacy of PBT. This study aimed to test the feasibility of a three-dimensional (3D) tumor-spheroid-based high-throughput screening platform that could assess cellular sensitivity against PBT. Spheroids of two HNSCC cell lines—Fadu and Cal27—cultured with a mixture of Matrigel were arrayed on a 384-pillar/well plate, followed by exposure to graded doses of protons or targeted drugs including olaparib at various concentrations. Calcein staining of HNSCC spheroids revealed a dose-dependent decrease in cell viability for proton irradiation or multiple targeted drugs, and provided quantitative data that discriminated the sensitivity between the two HNSCC cell lines. The combined effect of protons and olaparib was assessed by calculating the combination index from the survival rates of 4 × 4 matrices, showing that Cal27 spheroids had greater synergy with olaparib than Fadu spheroids. In contrast, adavosertib did not synergize with protons in both spheroids. Taken together, we demonstrated that the 3D pillar/well array platform was a useful tool that provided rapid, quantitative data for evaluating sensitivity to PBT and drug combinations. Our results further supported that administration of the combination of PBT and olaparib may be an effective treatment strategy for HNSCC patients.
Collapse
|
13
|
Paganetti H. Mechanisms and Review of Clinical Evidence of Variations in Relative Biological Effectiveness in Proton Therapy. Int J Radiat Oncol Biol Phys 2022; 112:222-236. [PMID: 34407443 PMCID: PMC8688199 DOI: 10.1016/j.ijrobp.2021.08.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/14/2021] [Accepted: 08/10/2021] [Indexed: 01/03/2023]
Abstract
Proton therapy is increasingly being used as a radiation therapy modality. There is uncertainty about the biological effectiveness of protons relative to photon therapies as it depends on several physical and biological parameters. Radiation oncology currently applies a constant and generic value for the relative biological effectiveness (RBE) of 1.1, which was chosen conservatively to ensure tumor coverage. The use of a constant value has been challenged particularly when considering normal tissue constraints. Potential variations in RBE have been assessed in several published reviews but have mostly focused on data from clonogenic cell survival experiments with unclear relevance for clinical proton therapy. The goal of this review is to put in vitro findings in relation to clinical observations. Relevant in vivo pathways determining RBE for tumors and normal tissues are outlined, including not only damage to tumor cells and parenchyma but also vascular damage and immune response. Furthermore, the current clinical evidence of varying RBE is reviewed. The assessment can serve as guidance for treatment planning, personalized dose prescriptions, and outcome analysis.
Collapse
Affiliation(s)
- Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
14
|
Du J, Kageyama SI, Hirata H, Motegi A, Nakamura M, Hirano Y, Okumura M, Yamashita R, Tsuchihara K, Hojo H, Hirayama R, Akimoto T. Comparative analysis of the immune responses in cancer cells irradiated with X-ray, proton and carbon-ion beams. Biochem Biophys Res Commun 2021; 585:55-60. [PMID: 34784552 DOI: 10.1016/j.bbrc.2021.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022]
Abstract
Radiotherapy (RT) is an effective treatment option for cancer; however, its efficacy remains less than optimal in locally advanced cancer. Immune checkpoint inhibitor-based therapy, including the administration of anti-PD-L1 antibodies, is a promising approach that works synergistically with RT. Proton beam therapy and carbon-ion therapy are common options for patients with cancer. Proton and carbon ions are reported to induce an immune reaction in cancer cells; however, the underlying mechanisms remain unclear. Here, we aimed to compare the immune responses after irradiation (IR) with X-ray, protons, and carbon ions in an oesophageal cancer cell line and the underlying mechanisms. An oesophageal cancer cell line, KYSE450, was irradiated with 1 fraction/15 GyE (Gy equivalent) of X-ray, proton, or carbon-ion beams, and then, the cells were harvested for RNA sequencing and gene enrichment analysis. We also knocked out STING and STAT1 in the quest for mechanistic insights. RNA sequencing data revealed that gene expression signatures and biological processes were different in KYSE450 irradiated with X-ray, proton, and carbon-ion beams 6-24 h after IR. However, after 3 days, a common gene expression signature was detected, associated with biological pathways involved in innate immune responses. Gene knock-out experiments revealed that the STING-STAT1 axis underlies the immune reactions after IR. X-Ray, proton, and carbon-ion IRs induced similar immune responses, regulated by the STING-STAT1 axis.
Collapse
Affiliation(s)
- Junyan Du
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Hidenari Hirata
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Atsushi Motegi
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Masaki Nakamura
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Yasuhiro Hirano
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Masayuki Okumura
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Hidehiro Hojo
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan.
| | - Ryoichi Hirayama
- Radiation and Cancer Biology Group, Department of Charged Particle Therapy Research, Institute for Quantum Medical Science (iQMS), Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology (QST), Chiba, Japan
| | - Tetsuo Akimoto
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Chiba, Japan; Department of Radiation Oncology, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
15
|
Matsumoto Y, Fukumitsu N, Ishikawa H, Nakai K, Sakurai H. A Critical Review of Radiation Therapy: From Particle Beam Therapy (Proton, Carbon, and BNCT) to Beyond. J Pers Med 2021; 11:jpm11080825. [PMID: 34442469 PMCID: PMC8399040 DOI: 10.3390/jpm11080825] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/20/2021] [Accepted: 08/22/2021] [Indexed: 12/24/2022] Open
Abstract
In this paper, we discuss the role of particle therapy—a novel radiation therapy (RT) that has shown rapid progress and widespread use in recent years—in multidisciplinary treatment. Three types of particle therapies are currently used for cancer treatment: proton beam therapy (PBT), carbon-ion beam therapy (CIBT), and boron neutron capture therapy (BNCT). PBT and CIBT have been reported to have excellent therapeutic results owing to the physical characteristics of their Bragg peaks. Variable drug therapies, such as chemotherapy, hormone therapy, and immunotherapy, are combined in various treatment strategies, and treatment effects have been improved. BNCT has a high dose concentration for cancer in terms of nuclear reactions with boron. BNCT is a next-generation RT that can achieve cancer cell-selective therapeutic effects, and its effectiveness strongly depends on the selective 10B accumulation in cancer cells by concomitant boron preparation. Therefore, drug delivery research, including nanoparticles, is highly desirable. In this review, we introduce both clinical and basic aspects of particle beam therapy from the perspective of multidisciplinary treatment, which is expected to expand further in the future.
Collapse
Affiliation(s)
- Yoshitaka Matsumoto
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
- Correspondence: ; Tel.: +81-29-853-7100
| | | | - Hitoshi Ishikawa
- National Institute of Quantum and Radiological Science and Technology Hospital, Chiba 263-8555, Japan;
| | - Kei Nakai
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
| | - Hideyuki Sakurai
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; (K.N.); (H.S.)
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan
| |
Collapse
|
16
|
Maccaroni E, Giampieri R, Lenci E, Scortichini L, Bianchi F, Belvederesi L, Brugiati C, Pagliaretta S, Ambrosini E, Berardi R. BRCA mutations and gastrointestinal cancers: When to expect the unexpected? World J Clin Oncol 2021; 12:565-580. [PMID: 34367929 PMCID: PMC8317649 DOI: 10.5306/wjco.v12.i7.565] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/17/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
BRCA1/2 pathogenic variants are widely known as major risk factors mainly for breast and ovarian cancer, while their role in gastrointestinal (GI) malignancies such as colorectal cancer (CRC), gastric cancer and oesophageal cancer (OeC) is still not well established. The main objective of this review is to summarise the available evidence on this matter. The studies included in the review were selected from PubMed/GoogleScholar/ScienceDirect databases to identify published articles where BRCA1/2 pathogenic variants were assessed either as a risk factor or a prognostic/predictive factor in these malignancies. Our review suggests that BRCA1/2 might have a role as a risk factor for colorectal, gastric and OeC, albeit with differences among these diseases: In particular BRCA1 seems to be much more frequently mutated in CRC whereas BRCA2 appears to be much more closely associated with gastric and OeC. Early-onset cancer seems to be also associated with BRCA1/2 mutations and a few studies suggest a positive prognostic role of these mutations. The assessment of a potentially predictive role of these mutations is hampered by the fact that most patients with these diseases have been treated with platinum compounds, where it is expected that a higher probability of response should be seen. A few clinical trials focused on poly (ADP-ribose) polymerase inhibitors use in GI cancers are currently ongoing.
Collapse
Affiliation(s)
- Elena Maccaroni
- Department of Oncology, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona 60126, Italy
| | - Riccardo Giampieri
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona 60126, Italy
| | - Edoardo Lenci
- Department of Oncology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Laura Scortichini
- Department of Oncology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Francesca Bianchi
- Molecular and Clinical Science Department, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Laura Belvederesi
- Molecular and Clinical Science Department, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Cristiana Brugiati
- Molecular and Clinical Science Department, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Silvia Pagliaretta
- Molecular and Clinical Science Department, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Elisa Ambrosini
- Molecular and Clinical Science Department, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Rossana Berardi
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti di Ancona, Ancona 60126, Italy
| |
Collapse
|
17
|
Rosenbaum MW, Gonzalez RS. Targeted therapy for upper gastrointestinal tract cancer: current and future prospects. Histopathology 2021; 78:148-161. [PMID: 33382497 DOI: 10.1111/his.14244] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric and oesophageal carcinoma remain major causes of worldwide mortality and morbidity. Despite incredible progress in understanding tumour biology, few targeted treatment options have proved effective in prolonging survival, and adjuvant therapy is largely interchangeable in these carcinomas. Through large-scale sequencing by the Cancer Genome Atlas and the Asian Cancer Research Group, numerous potential molecular targets have been discovered. Of the approved targeted therapies for gastric and oesophageal cancer, pathologists play a role in patient selection for the majority of them. Trastuzumab has been approved as a first-line therapy in conjunction with standard treatment in adenocarcinomas with either 3+ HER2/neu expression by immunohistochemistry or ERBB2 amplification by FISH. PD-L1 immunohistochemistry showing a combined positive score of 1 or greater qualifies patients for third-line pembrolizumab therapy, and identification of microsatellite instability-high carcinomas may qualify patients for second-line pembrolizumab. Ramucirumab, targeting VEGFR2, has also been approved for second-line therapy in gastric carcinoma. Non-surgical therapy for gastrointestinal stromal tumours relies mainly upon tyrosine kinase inhibitors, while new targeted therapy options for neuroendocrine neoplasms have recently emerged. Potential future options for targeted therapy in all these malignancies are being investigated in clinical trials, as this review will discuss.
Collapse
Affiliation(s)
- Matthew W Rosenbaum
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Raul S Gonzalez
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
18
|
Principe DR, Narbutis M, Koch R, Rana A. Frequency and prognostic value of mutations associated with the homologous recombination DNA repair pathway in a large pan cancer cohort. Sci Rep 2020; 10:20223. [PMID: 33214570 PMCID: PMC7677533 DOI: 10.1038/s41598-020-76975-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/27/2020] [Indexed: 12/18/2022] Open
Abstract
PARP inhibitors have shown remarkable efficacy in the clinical management of several BRCA-mutated tumors. This approach is based on the long-standing hypothesis that PARP inhibition will impair the repair of single stranded breaks, causing synthetic lethality in tumors with loss of high-fidelity double-strand break homologous recombination. While this is now well accepted and has been the basis of several successful clinical trials, emerging evidence strongly suggests that mutation to several additional genes involved in homologous recombination may also have predictive value for PARP inhibitors. While this notion is supported by early clinical evidence, the mutation frequencies of these and other functionally related genes are largely unknown, particularly in cancers not classically associated with homologous recombination deficiency. We therefore evaluated the mutation status of 22 genes associated with the homologous recombination DNA repair pathway or PARP inhibitor sensitivity, first in a pan-cancer cohort of 55,586 patients, followed by a more focused analysis in The Cancer Genome Atlas cohort of 12,153 patients. In both groups we observed high rates of mutations in a variety of HR-associated genes largely unexplored in the setting of PARP inhibition, many of which were associated also with poor clinical outcomes. We then extended our study to determine which mutations have a known oncogenic role, as well as similar to known oncogenic mutations that may have a similar phenotype. Finally, we explored the individual cancer histologies in which these genomic alterations are most frequent. We concluded that the rates of deleterious mutations affecting genes associated with the homologous recombination pathway may be underrepresented in a wide range of human cancers, and several of these genes warrant further and more focused investigation, particularly in the setting of PARP inhibition and HR deficiency.
Collapse
Affiliation(s)
- Daniel R Principe
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, IL, USA
| | - Matthew Narbutis
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA
| | - Regina Koch
- University of Illinois College of Medicine, Chicago, IL, USA
| | - Ajay Rana
- Division of Surgical Oncology, Department of Surgery, College of Medicine, The University of Illinois at Chicago, 840 S. Wood Street, Suite 601 Clinical Sciences Building, Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
| |
Collapse
|
19
|
Görte J, Beyreuther E, Danen EHJ, Cordes N. Comparative Proton and Photon Irradiation Combined with Pharmacological Inhibitors in 3D Pancreatic Cancer Cultures. Cancers (Basel) 2020; 12:cancers12113216. [PMID: 33142778 PMCID: PMC7692858 DOI: 10.3390/cancers12113216] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Due to higher precision and consequent sparing of normal tissue, pancreatic cancer patients might profit from proton beam radiotherapy, a treatment modality increasingly used. Since molecular data upon proton irradiation in comparison to standard photon radiotherapy are limited in pancreatic cancer, the aims of our study were to unravel differences in the effectiveness of photon versus proton irradiation and to exploit radiation type-specific molecular changes for radiosensitizing 3D PDAC cell cultures. Although protons showed a slightly higher effectiveness and a stronger induction of molecular alterations than photons, our results revealed a radiation-type independent sensitization of molecular-targeted agents selected according to the discovered molecular, radiation-induced alterations. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly therapy-resistant tumor entity of unmet needs. Over the last decades, radiotherapy has been considered as an additional treatment modality to surgery and chemotherapy. Owing to radiosensitive abdominal organs, high-precision proton beam radiotherapy has been regarded as superior to photon radiotherapy. To further elucidate the potential of combination therapies, we employed a more physiological 3D, matrix-based cell culture model to assess tumoroid formation capacity after photon and proton irradiation. Additionally, we investigated proton- and photon-irradiation-induced phosphoproteomic changes for identifying clinically exploitable targets. Here, we show that proton irradiation elicits a higher efficacy to reduce 3D PDAC tumoroid formation and a greater extent of phosphoproteome alterations compared with photon irradiation. The targeting of proteins identified in the phosphoproteome that were uniquely altered by protons or photons failed to cause radiation-type-specific radiosensitization. Targeting DNA repair proteins associated with non-homologous endjoining, however, revealed a strong radiosensitizing potential independent of the radiation type. In conclusion, our findings suggest proton irradiation to be potentially more effective in PDAC than photons without additional efficacy when combined with DNA repair inhibitors.
Collapse
Affiliation(s)
- Josephine Görte
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiation Physics, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Erik H. J. Danen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333CC Leiden, The Netherlands;
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus Technische Universität Dresden, 01307 Dresden, Germany; (J.G.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden: German Cancer Research Center, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Correspondence: ; Tel.: +49-351-458-7401; Fax: +49-351-458-7311
| |
Collapse
|