Could Protons and Carbon Ions Be the Silver Bullets Against Pancreatic Cancer?
Abstract
:1. Introduction
2. Current Treatment for Unmetastasised PDAC
3. From Adjuvant to Neoadjuvant Treatment For Unmetastasised PDAC
4. Pushing Forward Loco-Regional Control: Modern RT
4.1. Dose Conformation Towards Dose Escalation and Hypofractionation
4.2. Charged Particles to Increase Dose Response and Counteract PDAC Resistance to Treatment
4.2.1. Mechanisms of Resistance
4.2.2. Charged Particles vs. Resistance
4.3. Pushing Further PDAC Local Control: Charged Particles and Targeted Drug Combination
5. PDAC Systemic management: Charged Particles to Trigger an Immune Response
5.1. Immune Evasion in PDAC: T Cells and Tumour-Associated Macrophages
5.2. Conventional RT and Immune Response in PDAC
5.3. Charged Particles to Improve Immune Response in PDAC
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
3DCRT | three-dimensional conformation radiation therapy |
5-FU | 5-fluorouracil |
ABCG2 | ATP-binding cassette super-family G member 2 |
AKT | protein kinase B |
Arg | arginase |
ATM | ataxia telangiectasia mutated |
ATP | adenosine triphosphate |
BED | biological equivalent dose |
BRCA2 | breast cancer type 2 susceptibility protein |
BR-PDAC | borderline resectable PDAC |
CAF | cancer associated fibroblast |
CCL2 | chemokine ligand 2 |
CCR2 | C-C chemokine receptor type 2 |
CDK4/6 | cyclin-dependent kinase 4 and 6 |
CDKN2A | cyclin-dependent kinase inhibitor 2A |
cGAS | cyclic GMP-AMP synthase |
CRT | chemoradiotherapy |
CSC | cancer stem cell |
CSF1 | colony-stimulating factor |
CSF1R | colony-stimulating factor 1 receptor |
CT | chemotherapy |
CTLA4 | cytotoxic T-lymphocyte associated protein 4 |
CXCR4 | C-X-C motif chemokine receptor 4 |
DAMPs | damage associated molecular patterns |
DC | denditric cell |
EGFR | epidermal growth factor receptor |
EMT | epithelial to mesenchymal transition |
EORTC | European organisation for research and treatment of cancer |
ERK | mitogen-activated protein kinase 1 |
ESPAC | European study group for pancreatic cancer |
EV | extracellular vesicle |
FLASH RT | ultrahigh dose-rate radiation therapy |
GIST | gastrointestinal study group |
HIF-1α | hypoxia inducible factor 1 |
HMGB-1 | high mobility group box 1 |
HR | homologous recombination |
ICD | immunogenic cell death |
IFN-γ | interferon-γ |
IMRT | intensity modulated radiation therapy |
iNOS | inducible nitric oxide synthase |
KRAS | kirsten rat sarcoma |
LA-PDAC | locally advanced PDAC |
LET | linear energy transfer |
LPS | lipopolysaccharide |
MDSC | myeloid-derived suppressor cells |
MEK | mitogen-activated protein kinase kinase |
MHC 1 | major histocompatibility complex class 1 |
mTOR | mammalian target of rapamycin |
nab-paclitaxel | nanoparticle-bound paclitaxel |
NF-κB | nuclear factor kappa B |
NHEJ | non homologous end joining |
NRF2 | nuclear factor erythroid 2-related factor 2 |
OAR | organ at risk |
OS | overall survival |
PARP | poly(ADP-ribose) polymerase |
PDAC | pancreatic ductal adenocarcinoma |
PD1 | programmed cell death 1 |
PDK1 | pyruvate dehydrogenase kinase 1 |
PD-L1 | programmed cell death ligand 1 |
PDX | patient-derived xenograft |
PI3K | phosphatidylinositol 3-kinase |
POLO | pancreas cancer olaparib ongoing |
PREOPANC | preoperative chemoradiotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer |
PRODIGE | trial comparing adjuvant chemotherapy with gemcitabine versus mfolfirinox to treat resected pancreatic adenocarcinoma |
PT | proton therapy |
RAF | rapidly accelerated fibrosarcoma |
RAS | rat sarcoma |
RBE | relative biological effectiveness |
ROS | reactive oxygen species |
R-PDAC | resectable PDAC |
RT | radiotherapy |
RTOG | radiation therapy oncology group |
SBRT | stereotactic body radiation therapy |
SDF-1 | stromal derived factor 1 |
SMAD4 | mothers against decapentaplegic homolog 4 |
SMART | stereotactic magnetic resonance guide adaption radiation therapy |
SPArc | continuous spot-scanning protons arc |
STING | stimulator of interferon genes |
TAM | tumour-associated macrophages |
TGFβ | transforming growth factor beta |
TIL | tumour infiltrating lymphocytes |
TNF | tumour necrosis factor |
TRAIL | TNF-related apoptosis-inducing ligand |
Trex1 | three prime repair exonuclease 1 |
UPR | unfolded protein response |
VEGF | vascular endothelial growth factor |
Wnt | wingless-integration-1 |
ZEB | zinc finger E-box binding homeobox |
References
- Ferlay, J.; Colombet, M.; Soerjomataram, I.; Dyba, T.; Randi, G.; Bettio, M.; Gavin, A.; Visser, O.; Bray, F. Cancer incidence and mortality patterns in europe: Estimates for 40 countries and 25 major cancers in 2018. Eur. J. Cancer 2018, 103, 356–387. [Google Scholar] [CrossRef]
- Kleeff, J.; Korc, M.; Apte, M.; La Vecchia, C.; Johnson, C.D.; Biankin, A.V.; Neale, R.E.; Tempero, M.; Tuveson, D.A.; Hruban, R.H.; et al. Pancreatic cancer. Nat. Rev. Dis. Primers 2016, 2, 16022. [Google Scholar] [CrossRef]
- Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 2014, 371, 2140–2141. [Google Scholar] [CrossRef]
- Ducreux, M.; Seufferlein, T.; Van Laethem, J.L.; Laurent-Puig, P.; Smolenschi, C.; Malka, D.; Boige, V.; Hollebecque, A.; Conroy, T. Systemic treatment of pancreatic cancer revisited. Semin. Oncol. 2019, 46, 28–38. [Google Scholar] [CrossRef] [PubMed]
- Neoptolemos, J.P.; Kleeff, J.; Michl, P.; Costello, E.; Greenhalf, W.; Palmer, D.H. Therapeutic developments in pancreatic cancer: Current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 333–348. [Google Scholar] [CrossRef] [PubMed]
- Conroy, T.; Gavoille, C.; Samalin, E.; Ychou, M.; Ducreux, M. The role of the FOLFIRINOX regimen for advanced pancreatic cancer. Curr. Oncol. Rep. 2013, 15, 182–189. [Google Scholar] [CrossRef] [PubMed]
- Amrutkar, M.; Gladhaug, I.P. Pancreatic Cancer Chemoresistance to Gemcitabine. Cancers 2017, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Sousa Cavalcante, L.; Monteiro, G. Gemcitabine: Metabolism and molecular mechanisms of action, sensitivity and chemoresistance in pancreatic cancer. Eur. J. Pharmacol. 2014, 741, 8–16. [Google Scholar] [CrossRef] [PubMed]
- Neoptolemos, J.P.; Palmer, D.H.; Ghaneh, P.; Psarelli, E.E.; Valle, J.W.; Halloran, C.M.; Faluyi, O.; O’Reilly, D.A.; Cunningham, D.; Wadsley, J.; et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet 2017, 389, 1011–1024. [Google Scholar] [CrossRef]
- Springfeld, C.; Jager, D.; Buchler, M.W.; Strobel, O.; Hackert, T.; Palmer, D.H.; Neoptolemos, J.P. Chemotherapy for pancreatic cancer. Presse Med. 2019, 48, e159–e174. [Google Scholar] [CrossRef]
- Hall, W.A.; Goodman, K.A. Radiation therapy for pancreatic adenocarcinoma, a treatment option that must be considered in the management of a devastating malignancy. Radiat. Oncol. 2019, 14, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- GIST. Further evidence of effective adjuvant combined radiation and chemotherapy following curative resection of pancreatic cancer. Cancer 1987, 59, 2006–2010. [Google Scholar] [CrossRef]
- Klinkenbijl, J.H.; Jeekel, J.; Sahmoud, T.; van Pel, R.; Couvreur, M.L.; Veenhof, C.H.; Arnaud, J.P.; Gonzalez, D.G.; de Wit, L.T.; Hennipman, A.; et al. Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: Phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann. Surg. 1999, 230, 776–782, discussion 782–774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Neoptolemos, J.P.; Stocken, D.D.; Friess, H.; Bassi, C.; Dunn, J.A.; Hickey, H.; Beger, H.; Fernandez-Cruz, L.; Dervenis, C.; Lacaine, F.; et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N. Engl. J. Med. 2004, 350, 1200–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Regine, W.F.; Winter, K.A.; Abrams, R.; Safran, H.; Hoffman, J.P.; Konski, A.; Benson, A.B.; Macdonald, J.S.; Rich, T.A.; Willett, C.G. Fluorouracil-based chemoradiation with either gemcitabine or fluorouracil chemotherapy after resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. Intergroup/RTOG 9704 phase III trial. Ann. Surg. Oncol. 2011, 18, 1319–1326. [Google Scholar] [CrossRef] [Green Version]
- Abrams, R.A.; Winter, K.A.; Regine, W.F.; Safran, H.; Hoffman, J.P.; Lustig, R.; Konski, A.A.; Benson, A.B.; Macdonald, J.S.; Rich, T.A.; et al. Failure to adhere to protocol specified radiation therapy guidelines was associated with decreased survival in RTOG 9704—A phase III trial of adjuvant chemotherapy and chemoradiotherapy for patients with resected adenocarcinoma of the pancreas. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 809–816. [Google Scholar] [CrossRef] [Green Version]
- Konstantinidis, I.T.; Warshaw, A.L.; Allen, J.N.; Blaszkowsky, L.S.; Castillo, C.F.; Deshpande, V.; Hong, T.S.; Kwak, E.L.; Lauwers, G.Y.; Ryan, D.P.; et al. Pancreatic ductal adenocarcinoma: Is there a survival difference for R1 resections versus locally advanced unresectable tumors? What is a “true” R0 resection? Ann. Surg. 2013, 257, 731–736. [Google Scholar] [CrossRef]
- Versteijne, E.; Suker, M.; Groothuis, K.; Akkermans-Vogelaar, J.M.; Besselink, M.G.; Bonsing, B.A.; Buijsen, J.; Busch, O.R.; Creemers, G.M.; van Dam, R.M.; et al. Preoperative Chemoradiotherapy Versus Immediate Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Results of the Dutch Randomized Phase III PREOPANC Trial. J. Clin. Oncol. 2020, 38, 1763–1773. [Google Scholar] [CrossRef]
- Janssen, Q.P.; Buettner, S.; Suker, M.; Beumer, B.R.; Addeo, P.; Bachellier, P.; Bahary, N.; Bekaii-Saab, T.; Bali, M.A.; Besselink, M.G.; et al. Neoadjuvant FOLFIRINOX in Patients With Borderline Resectable Pancreatic Cancer: A Systematic Review and Patient-Level Meta-Analysis. J. Natl. Cancer Inst. 2019, 111, 782–794. [Google Scholar] [CrossRef]
- Janssen, Q.P.; O’Reilly, E.M.; van Eijck, C.H.J.; Groot Koerkamp, B. Neoadjuvant Treatment in Patients With Resectable and Borderline Resectable Pancreatic Cancer. Front. Oncol. 2020, 10, 41. [Google Scholar] [CrossRef]
- Dhir, M.; Zenati, M.S.; Hamad, A.; Singhi, A.D.; Bahary, N.; Hogg, M.E.; Zeh, H.J., 3rd; Zureikat, A.H. FOLFIRINOX Versus Gemcitabine/Nab-Paclitaxel for Neoadjuvant Treatment of Resectable and Borderline Resectable Pancreatic Head Adenocarcinoma. Ann. Surg. Oncol. 2018, 25, 1896–1903. [Google Scholar] [CrossRef] [PubMed]
- Klaiber, U.; Hackert, T. Conversion Surgery for Pancreatic Cancer-The Impact of Neoadjuvant Treatment. Front. Oncol. 2019, 9, 1501. [Google Scholar] [CrossRef] [Green Version]
- Gillen, S.; Schuster, T.; Meyer Zum Buschenfelde, C.; Friess, H.; Kleeff, J. Preoperative/neoadjuvant therapy in pancreatic cancer: A systematic review and meta-analysis of response and resection percentages. PLoS Med. 2010, 7, e1000267. [Google Scholar] [CrossRef] [Green Version]
- Bittner, M.I.; Grosu, A.L.; Brunner, T.B. Comparison of toxicity after IMRT and 3D-conformal radiotherapy for patients with pancreatic cancer—A systematic review. Radiother. Oncol. 2015, 114, 117–121. [Google Scholar] [CrossRef]
- Krishnan, S.; Chadha, A.S.; Suh, Y.; Chen, H.C.; Rao, A.; Das, P.; Minsky, B.D.; Mahmood, U.; Delclos, M.E.; Sawakuchi, G.O.; et al. Focal Radiation Therapy Dose Escalation Improves Overall Survival in Locally Advanced Pancreatic Cancer Patients Receiving Induction Chemotherapy and Consolidative Chemoradiation. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 755–765. [Google Scholar] [CrossRef] [Green Version]
- Reyngold, M.; Parikh, P.; Crane, C.H. Ablative radiation therapy for locally advanced pancreatic cancer: Techniques and results. Radiat. Oncol. 2019, 14, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ling, T.C.; Slater, J.M.; Mifflin, R.; Nookala, P.; Grove, R.; Ly, A.M.; Patyal, B.; Slater, J.D.; Yang, G.Y. Evaluation of normal tissue exposure in patients receiving radiotherapy for pancreatic cancer based on RTOG 0848. J. Gastrointest. Oncol. 2015, 6, 108–114. [Google Scholar] [CrossRef] [PubMed]
- Rutenberg, M.S.; Nichols, R.C. Proton beam radiotherapy for pancreas cancer. J. Gastrointest. Oncol. 2020, 11, 166–175. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.H.; Lee, W.J.; Woo, S.M.; Kim, H.; Oh, E.S.; Lee, J.H.; Han, S.S.; Park, S.J.; Suh, Y.G.; Moon, S.H.; et al. Effectiveness and Safety of Simultaneous Integrated Boost-Proton Beam Therapy for Localized Pancreatic Cancer. Technol. Cancer Res. Treat. 2018, 17, 1533033818783879. [Google Scholar] [CrossRef]
- Hiroshima, Y.; Fukumitsu, N.; Saito, T.; Numajiri, H.; Murofushi, K.N.; Ohnishi, K.; Nonaka, T.; Ishikawa, H.; Okumura, T.; Sakurai, H. Concurrent chemoradiotherapy using proton beams for unresectable locally advanced pancreatic cancer. Radiother. Oncol. 2019, 136, 37–43. [Google Scholar] [CrossRef] [Green Version]
- Ding, X.; Li, X.; Qin, A.; Zhou, J.; Yan, D.; Stevens, C.; Krauss, D.; Kabolizdeh, P. Have we reached proton beam therapy dosimetric limitations?-A novel robust, delivery-efficient and continuous spot-scanning proton arc (SPArc) therapy is to improve the dosimetric outcome in treating prostate cancer. Acta Oncol. 2018, 57, 435–437. [Google Scholar] [CrossRef] [PubMed]
- Schreuder, A.N.; Shamblin, J. Proton therapy delivery: What is needed in the next ten years? Br. J. Radiol. 2020, 93, 20190359. [Google Scholar] [CrossRef] [PubMed]
- Shinoto, M.; Yamada, S.; Terashima, K.; Yasuda, S.; Shioyama, Y.; Honda, H.; Kamada, T.; Tsujii, H.; Saisho, H. Carbon Ion Radiation Therapy With Concurrent Gemcitabine for Patients With Locally Advanced Pancreatic Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 498–504. [Google Scholar] [CrossRef] [PubMed]
- Dell’Oro, M.; Short, M.; Wilson, P.; Bezak, E. Clinical Limitations of Photon, Proton and Carbon Ion Therapy for Pancreatic Cancer. Cancers 2020, 12, 163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vozenin, M.C.; Hendry, J.H.; Limoli, C.L. Biological Benefits of Ultra-high Dose Rate FLASH Radiotherapy: Sleeping Beauty Awoken. Clin. Oncol. 2019, 31, 407–415. [Google Scholar] [CrossRef] [PubMed]
- Wilson, J.D.; Hammond, E.M.; Higgins, G.S.; Petersson, K. Ultra-High Dose Rate (FLASH) Radiotherapy: Silver Bullet or Fool’s Gold? Front. Oncol. 2019, 9, 1563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Favaudon, V.; Caplier, L.; Monceau, V.; Pouzoulet, F.; Sayarath, M.; Fouillade, C.; Poupon, M.F.; Brito, I.; Hupe, P.; Bourhis, J.; et al. Ultrahigh dose-rate FLASH irradiation increases the differential response between normal and tumor tissue in mice. Sci. Transl. Med. 2014, 6, 245ra293. [Google Scholar] [CrossRef]
- Diffenderfer, E.S.; Verginadis, I.I.; Kim, M.M.; Shoniyozov, K.; Velalopoulou, A.; Goia, D.; Putt, M.; Hagan, S.; Avery, S.; Teo, K.; et al. Design, Implementation, and in Vivo Validation of a Novel Proton FLASH Radiation Therapy System. Int. J. Radiat. Oncol. Biol. Phys. 2020, 106, 440–448. [Google Scholar] [CrossRef] [Green Version]
- Bourhis, J.; Sozzi, W.J.; Jorge, P.G.; Gaide, O.; Bailat, C.; Duclos, F.; Patin, D.; Ozsahin, M.; Bochud, F.; Germond, J.F.; et al. Treatment of a first patient with FLASH-radiotherapy. Radiother. Oncol. 2019, 139, 18–22. [Google Scholar] [CrossRef]
- Seshacharyulu, P.; Baine, M.J.; Souchek, J.J.; Menning, M.; Kaur, S.; Yan, Y.; Ouellette, M.M.; Jain, M.; Lin, C.; Batra, S.K. Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 69–92. [Google Scholar] [CrossRef]
- Janes, M.R.; Zhang, J.; Li, L.S.; Hansen, R.; Peters, U.; Guo, X.; Chen, Y.; Babbar, A.; Firdaus, S.J.; Darjania, L.; et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell 2018, 172, 578–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fakih, M.; O’Neil, B.; Price, T.J.; Falchook, G.S.; Desai, J.; Kuo, J.; Govindan, R.; Rasmussen, E.; Morrow, P.K.H.; Ngang, J.; et al. Phase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of AMG 510, a novel small molecule KRASG12C inhibitor, in advanced solid tumors. J. Clin. Oncol. 2019, 37, 3003. [Google Scholar] [CrossRef]
- Kinsey, C.G.; Camolotto, S.A.; Boespflug, A.M.; Guillen, K.P.; Foth, M.; Truong, A.; Schuman, S.S.; Shea, J.E.; Seipp, M.T.; Yap, J.T.; et al. Protective autophagy elicited by RAF-->MEK-->ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat. Med. 2019, 25, 620–627. [Google Scholar] [CrossRef] [PubMed]
- Panieri, E.; Buha, A.; Telkoparan-Akillilar, P.; Cevik, D.; Kouretas, D.; Veskoukis, A.; Skaperda, Z.; Tsatsakis, A.; Wallace, D.; Suzen, S.; et al. Potential Applications of NRF2 Modulators in Cancer Therapy. Antioxidants 2020, 9, 193. [Google Scholar] [CrossRef] [Green Version]
- Bailleul, J.; Yazal, T.; Sung, D.; Palomera, D.; Sehgal, A.; Dao, A.; Vlashi, E. Abstract C59: NRF2 drives metabolic reprogramming in irradiated pancreatic cancer cells and promotes radioresistance. Cancer Res. 2019, 79, C59. [Google Scholar] [CrossRef]
- Gorgulu, K.; Diakopoulos, K.N.; Kaya-Aksoy, E.; Ciecielski, K.J.; Ai, J.; Lesina, M.; Algul, H. The Role of Autophagy in Pancreatic Cancer: From Bench to the Dark Bedside. Cells 2020, 9, 1063. [Google Scholar] [CrossRef]
- Wang, F.; Xia, X.; Yang, C.; Shen, J.; Mai, J.; Kim, H.C.; Kirui, D.; Kang, Y.; Fleming, J.B.; Koay, E.J.; et al. SMAD4 Gene Mutation Renders Pancreatic Cancer Resistance to Radiotherapy through Promotion of Autophagy. Clin. Cancer Res. 2018, 24, 3176–3185. [Google Scholar] [CrossRef] [Green Version]
- Bailey, P.; Chang, D.K.; Nones, K.; Johns, A.L.; Patch, A.M.; Gingras, M.C.; Miller, D.K.; Christ, A.N.; Bruxner, T.J.; Quinn, M.C.; et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016, 531, 47–52. [Google Scholar] [CrossRef]
- Salvador-Barbero, B.; Alvarez-Fernandez, M.; Zapatero-Solana, E.; El Bakkali, A.; Menendez, M.D.C.; Lopez-Casas, P.P.; Di Domenico, T.; Xie, T.; VanArsdale, T.; Shields, D.J.; et al. CDK4/6 Inhibitors Impair Recovery from Cytotoxic Chemotherapy in Pancreatic Adenocarcinoma. Cancer Cell 2020, 37, 340–353. [Google Scholar] [CrossRef]
- Ruscetti, M.; Morris, J.P.t.; Mezzadra, R.; Russell, J.; Leibold, J.; Romesser, P.B.; Simon, J.; Kulick, A.; Ho, Y.J.; Fennell, M.; et al. Senescence-Induced Vascular Remodeling Creates Therapeutic Vulnerabilities in Pancreas Cancer. Cell 2020, 181, 424–441. [Google Scholar] [CrossRef]
- Thomas, D.; Radhakrishnan, P. Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol. Cancer 2019, 18, 14. [Google Scholar] [CrossRef] [PubMed]
- Grasso, C.; Jansen, G.; Giovannetti, E. Drug resistance in pancreatic cancer: Impact of altered energy metabolism. Crit. Rev. Oncol. Hematol. 2017, 114, 139–152. [Google Scholar] [CrossRef] [PubMed]
- Michiels, C.; Tellier, C.; Feron, O. Cycling hypoxia: A key feature of the tumor microenvironment. Biochim. Biophys. Acta 2016, 1866, 76–86. [Google Scholar] [CrossRef] [PubMed]
- Ma, N.Y.; Tinganelli, W.; Maier, A.; Durante, M.; Kraft-Weyrather, W. Influence of chronic hypoxia and radiation quality on cell survival. J. Radiat. Res. 2013, 54 (Suppl. 1), i13–i22. [Google Scholar] [CrossRef]
- Zolzer, F.; Streffer, C. Increased radiosensitivity with chronic hypoxia in four human tumor cell lines. Int. J. Radiat. Oncol. Biol. Phys. 2002, 54, 910–920. [Google Scholar] [CrossRef]
- Rebucci, M.; Michiels, C. Molecular aspects of cancer cell resistance to chemotherapy. Biochem. Pharmacol. 2013, 85, 1219–1226. [Google Scholar] [CrossRef]
- Huang, R.; Zhou, P.-K. HIF-1 signaling: A key orchestrator of cancer radioresistance. Radiat. Med. Prot. 2020, 1, 7–14. [Google Scholar] [CrossRef]
- Yokoi, K.; Fidler, I.J. Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin. Cancer Res. 2004, 10, 2299–2306. [Google Scholar] [CrossRef] [Green Version]
- He, X.; Wang, J.; Wei, W.; Shi, M.; Xin, B.; Zhang, T.; Shen, X. Hypoxia regulates ABCG2 activity through the activivation of ERK1/2/HIF-1alpha and contributes to chemoresistance in pancreatic cancer cells. Cancer Biol. Ther. 2016, 17, 188–198. [Google Scholar] [CrossRef] [Green Version]
- Schwartz, D.L.; Bankson, J.A.; Lemos, R., Jr.; Lai, S.Y.; Thittai, A.K.; He, Y.; Hostetter, G.; Demeure, M.J.; Von Hoff, D.D.; Powis, G. Radiosensitization and stromal imaging response correlates for the HIF-1 inhibitor PX-478 given with or without chemotherapy in pancreatic cancer. Mol. Cancer Ther. 2010, 9, 2057–2067. [Google Scholar] [CrossRef] [Green Version]
- Connell, P.P.; Hellman, S. Advances in radiotherapy and implications for the next century: A historical perspective. Cancer Res. 2009, 69, 383–392. [Google Scholar] [CrossRef] [Green Version]
- Nahum, A.E. The radiobiology of hypofractionation. Clin. Oncol. 2015, 27, 260–269. [Google Scholar] [CrossRef] [PubMed]
- Chapman, J.D. Can the two mechanisms of tumor cell killing by radiation be exploited for therapeutic gain? J. Radiat. Res. 2014, 55, 2–9. [Google Scholar] [CrossRef] [PubMed]
- Sia, J.; Szmyd, R.; Hau, E.; Gee, H.E. Molecular Mechanisms of Radiation-Induced Cancer Cell Death: A Primer. Front. Cell Dev. Biol. 2020, 8, 41. [Google Scholar] [CrossRef] [PubMed]
- Karam, S.D.; Bhatia, S. The radiobiological targets of SBRT: Tumor cells or endothelial cells? Ann. Transl. Med. 2015, 3, 290. [Google Scholar] [CrossRef] [PubMed]
- Moding, E.J.; Mowery, Y.M.; Kirsch, D.G. Opportunities for Radiosensitization in the Stereotactic Body Radiation Therapy (SBRT) Era. Cancer J. 2016, 22, 267–273. [Google Scholar] [CrossRef] [Green Version]
- Atashzar, M.R.; Baharlou, R.; Karami, J.; Abdollahi, H.; Rezaei, R.; Pourramezan, F.; Zoljalali Moghaddam, S.H. Cancer stem cells: A review from origin to therapeutic implications. J. Cell. Physiol. 2020, 235, 790–803. [Google Scholar] [CrossRef]
- Wang, Z.; Li, Y.; Ahmad, A.; Banerjee, S.; Azmi, A.S.; Kong, D.; Sarkar, F.H. Pancreatic cancer: Understanding and overcoming chemoresistance. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 27–33. [Google Scholar] [CrossRef]
- Zhou, S.; Zhang, M.; Zhou, C.; Wang, W.; Yang, H.; Ye, W. The role of epithelial-mesenchymal transition in regulating radioresistance. Crit. Rev. Oncol. Hematol. 2020, 150, 102961. [Google Scholar] [CrossRef]
- Duong, H.Q.; Hwang, J.S.; Kim, H.J.; Kang, H.J.; Seong, Y.S.; Bae, I. Aldehyde dehydrogenase 1A1 confers intrinsic and acquired resistance to gemcitabine in human pancreatic adenocarcinoma MIA PaCa-2 cells. Int. J. Oncol. 2012, 41, 855–861. [Google Scholar] [CrossRef] [Green Version]
- Hong, S.P.; Wen, J.; Bang, S.; Park, S.; Song, S.Y. CD44-positive cells are responsible for gemcitabine resistance in pancreatic cancer cells. Int. J. Cancer J. Int. Du Cancer 2009, 125, 2323–2331. [Google Scholar] [CrossRef] [PubMed]
- Shah, A.N.; Summy, J.M.; Zhang, J.; Park, S.I.; Parikh, N.U.; Gallick, G.E. Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann. Surg. Oncol. 2007, 14, 3629–3637. [Google Scholar] [CrossRef] [PubMed]
- Hermann, P.C.; Huber, S.L.; Herrler, T.; Aicher, A.; Ellwart, J.W.; Guba, M.; Bruns, C.J.; Heeschen, C. Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell 2007, 1, 313–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Wei, Q.; Wang, X.; Tang, S.; Liu, H.; Zhang, F.; Mohammed, M.K.; Huang, J.; Guo, D.; Lu, M.; et al. Transition to resistance: An unexpected role of the EMT in cancer chemoresistance. Genes Dis. 2016, 3, 3–6. [Google Scholar] [CrossRef] [Green Version]
- Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [Green Version]
- Elaskalani, O.; Razak, N.B.; Falasca, M.; Metharom, P. Epithelial-mesenchymal transition as a therapeutic target for overcoming chemoresistance in pancreatic cancer. World J. Gastrointest. Oncol. 2017, 9, 37–41. [Google Scholar] [CrossRef]
- Arumugam, T.; Ramachandran, V.; Fournier, K.F.; Wang, H.; Marquis, L.; Abbruzzese, J.L.; Gallick, G.E.; Logsdon, C.D.; McConkey, D.J.; Choi, W. Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 2009, 69, 5820–5828. [Google Scholar] [CrossRef] [Green Version]
- Lambies, G.; Miceli, M.; Martinez-Guillamon, C.; Olivera-Salguero, R.; Pena, R.; Frias, C.P.; Calderon, I.; Atanassov, B.S.; Dent, S.Y.R.; Arribas, J.; et al. TGFbeta-Activated USP27X Deubiquitinase Regulates Cell Migration and Chemoresistance via Stabilization of Snail1. Cancer Res. 2019, 79, 33–46. [Google Scholar] [CrossRef] [Green Version]
- Lonardo, E.; Hermann, P.C.; Mueller, M.T.; Huber, S.; Balic, A.; Miranda-Lorenzo, I.; Zagorac, S.; Alcala, S.; Rodriguez-Arabaolaza, I.; Ramirez, J.C.; et al. Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell 2011, 9, 433–446. [Google Scholar] [CrossRef] [Green Version]
- Adamska, A.; Elaskalani, O.; Emmanouilidi, A.; Kim, M.; Abdol Razak, N.B.; Metharom, P.; Falasca, M. Molecular and cellular mechanisms of chemoresistance in pancreatic cancer. Adv. Biol. Regul. 2018, 68, 77–87. [Google Scholar] [CrossRef]
- Tsukasa, K.; Ding, Q.; Yoshimitsu, M.; Miyazaki, Y.; Matsubara, S.; Takao, S. Slug contributes to gemcitabine resistance through epithelial-mesenchymal transition in CD133(+) pancreatic cancer cells. Hum. Cell 2015, 28, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Perusina Lanfranca, M.; Thompson, J.K.; Bednar, F.; Halbrook, C.; Lyssiotis, C.; Levi, B.; Frankel, T.L. Metabolism and epigenetics of pancreatic cancer stem cells. Semin. Cancer Biol. 2019, 57, 19–26. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Shi, P.; Zhao, G.; Xu, J.; Peng, W.; Zhang, J.; Zhang, G.; Wang, X.; Dong, Z.; Chen, F.; et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct. Target. Ther. 2020, 5, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cioffi, M.; Trabulo, S.M.; Sanchez-Ripoll, Y.; Miranda-Lorenzo, I.; Lonardo, E.; Dorado, J.; Reis Vieira, C.; Ramirez, J.C.; Hidalgo, M.; Aicher, A.; et al. The miR-17-92 cluster counteracts quiescence and chemoresistance in a distinct subpopulation of pancreatic cancer stem cells. Gut 2015, 64, 1936–1948. [Google Scholar] [CrossRef]
- Cheng, Z.X.; Wang, D.W.; Liu, T.; Liu, W.X.; Xia, W.B.; Xu, J.; Zhang, Y.H.; Qu, Y.K.; Guo, L.Q.; Ding, L.; et al. Effects of the HIF-1alpha and NF-kappaB loop on epithelial-mesenchymal transition and chemoresistance induced by hypoxia in pancreatic cancer cells. Oncol. Rep. 2014, 31, 1891–1898. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Han, H.; Rong, Y.; Zhu, K.; Zhu, Z.; Tang, Z.; Xiong, C.; Tao, J. Hypoxia potentiates gemcitabine-induced stemness in pancreatic cancer cells through AKT/Notch1 signaling. J. Exp. Clin. Cancer Res. 2018, 37, 291. [Google Scholar] [CrossRef]
- Khan, M.A.; Zubair, H.; Srivastava, S.K.; Singh, S.; Singh, A.P. Insights into the Role of microRNAs in Pancreatic Cancer Pathogenesis: Potential for Diagnosis, Prognosis, and Therapy. Adv. Exp. Med. Biol. 2015, 889, 71–87. [Google Scholar] [CrossRef] [Green Version]
- Ren, L.; Yu, Y. The role of miRNAs in the diagnosis, chemoresistance, and prognosis of pancreatic ductal adenocarcinoma. Ther. Clin. Risk Manag. 2018, 14, 179–187. [Google Scholar] [CrossRef] [Green Version]
- Gandellini, P.; Rancati, T.; Valdagni, R.; Zaffaroni, N. miRNAs in tumor radiation response: Bystanders or participants? Trends Mol. Med. 2014, 20, 529–539. [Google Scholar] [CrossRef]
- Liu, B.; Wu, S.; Ma, J.; Yan, S.; Xiao, Z.; Wan, L.; Zhang, F.; Shang, M.; Mao, A. lncRNA GAS5 Reverses EMT and Tumor Stem Cell-Mediated Gemcitabine Resistance and Metastasis by Targeting miR-221/SOCS3 in Pancreatic Cancer. Mol. Ther. Nucleic Acids 2018, 13, 472–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hwang, J.H.; Voortman, J.; Giovannetti, E.; Steinberg, S.M.; Leon, L.G.; Kim, Y.T.; Funel, N.; Park, J.K.; Kim, M.A.; Kang, G.H.; et al. Identification of microRNA-21 as a biomarker for chemoresistance and clinical outcome following adjuvant therapy in resectable pancreatic cancer. PLoS ONE 2010, 5, e10630. [Google Scholar] [CrossRef]
- Moriyama, T.; Ohuchida, K.; Mizumoto, K.; Yu, J.; Sato, N.; Nabae, T.; Takahata, S.; Toma, H.; Nagai, E.; Tanaka, M. MicroRNA-21 modulates biological functions of pancreatic cancer cells including their proliferation, invasion, and chemoresistance. Mol. Cancer Ther. 2009, 8, 1067–1074. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Ren, D.; Wu, X.; Lin, X.; Ye, L.; Lin, C.; Wu, S.; Zhu, J.; Peng, X.; Song, L. miR-1266 Contributes to Pancreatic Cancer Progression and Chemoresistance by the STAT3 and NF-kappaB Signaling Pathways. Mol. Ther. Nucleic Acids 2018, 11, 142–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, P.; Liu, L.; Yin, Y.; Zhao, Z.; Zhang, Y.; Amponsah, P.S.; Xiao, X.; Bauer, N.; Abukiwan, A.; Nwaeburu, C.C.; et al. MicroRNA-101-3p reverses gemcitabine resistance by inhibition of ribonucleotide reductase M1 in pancreatic cancer. Cancer Lett. 2016, 373, 130–137. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; VandenBoom, T.G., 2nd; Kong, D.; Wang, Z.; Ali, S.; Philip, P.A.; Sarkar, F.H. Up-regulation of miR-200 and let-7 by natural agents leads to the reversal of epithelial-to-mesenchymal transition in gemcitabine-resistant pancreatic cancer cells. Cancer Res. 2009, 69, 6704–6712. [Google Scholar] [CrossRef] [Green Version]
- Nalls, D.; Tang, S.N.; Rodova, M.; Srivastava, R.K.; Shankar, S. Targeting epigenetic regulation of miR-34a for treatment of pancreatic cancer by inhibition of pancreatic cancer stem cells. PLoS ONE 2011, 6, e24099. [Google Scholar] [CrossRef]
- Fesler, A.; Ju, J. Development of microRNA-based therapy for pancreatic cancer. J. Pancreatol. 2019, 2, 147–151. [Google Scholar] [CrossRef]
- Wang, W.; Zhao, L.; Wei, X.; Wang, L.; Liu, S.; Yang, Y.; Wang, F.; Sun, G.; Zhang, J.; Ma, Y.; et al. MicroRNA-320a promotes 5-FU resistance in human pancreatic cancer cells. Sci. Rep. 2016, 6, 27641. [Google Scholar] [CrossRef]
- Zhao, L.; Zou, D.; Wei, X.; Wang, L.; Zhang, Y.; Liu, S.; Si, Y.; Zhao, H.; Wang, F.; Yu, J.; et al. MiRNA-221-3p desensitizes pancreatic cancer cells to 5-fluorouracil by targeting RB1. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2016. [Google Scholar] [CrossRef]
- Wang, S.-L.; Wang, Y.-J.; Wu, S.-M.; Yuan, Z.; Du, H.-M.; Liu, X.; Zhu, X.-K. Identification of unique microRNAs associated with pancreatic cancer radioresistance using deep sequencing. Transl. Cancer Res. 2018, 7, 828–838. [Google Scholar] [CrossRef]
- Zhang, X.; Shi, H.; Lin, S.; Ba, M.; Cui, S. MicroRNA-216a enhances the radiosensitivity of pancreatic cancer cells by inhibiting beclin-1-mediated autophagy. Oncol. Rep. 2015, 34, 1557–1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, P.; Zhang, J.; Zhang, L.; Zhu, Z.; Fan, J.; Chen, L.; Zhuang, L.; Luo, J.; Chen, H.; Liu, L.; et al. MicroRNA 23b regulates autophagy associated with radioresistance of pancreatic cancer cells. Gastroenterology 2013, 145, 1133–1143. [Google Scholar] [CrossRef] [PubMed]
- Maacha, S.; Bhat, A.A.; Jimenez, L.; Raza, A.; Haris, M.; Uddin, S.; Grivel, J.C. Extracellular vesicles-mediated intercellular communication: Roles in the tumor microenvironment and anti-cancer drug resistance. Mol. Cancer 2019, 18, 55. [Google Scholar] [CrossRef] [Green Version]
- Ni, J.; Bucci, J.; Malouf, D.; Knox, M.; Graham, P.; Li, Y. Exosomes in Cancer Radioresistance. Front. Oncol. 2019, 9, 869. [Google Scholar] [CrossRef] [Green Version]
- Lucchetti, D.; Ricciardi Tenore, C.; Colella, F.; Sgambato, A. Extracellular Vesicles and Cancer: A Focus on Metabolism, Cytokines, and Immunity. Cancers 2020, 12, 171. [Google Scholar] [CrossRef] [Green Version]
- O’Neill, C.P.; Gilligan, K.E.; Dwyer, R.M. Role of Extracellular Vesicles (EVs) in Cell Stress Response and Resistance to Cancer Therapy. Cancers 2019, 11, 136. [Google Scholar] [CrossRef] [Green Version]
- Mikamori, M.; Yamada, D.; Eguchi, H.; Hasegawa, S.; Kishimoto, T.; Tomimaru, Y.; Asaoka, T.; Noda, T.; Wada, H.; Kawamoto, K.; et al. MicroRNA-155 Controls Exosome Synthesis and Promotes Gemcitabine Resistance in Pancreatic Ductal Adenocarcinoma. Sci. Rep. 2017, 7, 42339. [Google Scholar] [CrossRef]
- Binenbaum, Y.; Fridman, E.; Yaari, Z.; Milman, N.; Schroeder, A.; Ben David, G.; Shlomi, T.; Gil, Z. Transfer of miRNA in Macrophage-Derived Exosomes Induces Drug Resistance in Pancreatic Adenocarcinoma. Cancer Res. 2018, 78, 5287–5299. [Google Scholar] [CrossRef] [Green Version]
- Richards, K.E.; Zeleniak, A.E.; Fishel, M.L.; Wu, J.; Littlepage, L.E.; Hill, R. Cancer-associated fibroblast exosomes regulate survival and proliferation of pancreatic cancer cells. Oncogene 2017, 36, 1770–1778. [Google Scholar] [CrossRef] [Green Version]
- Karger, C.P.; Peschke, P. RBE and related modeling in carbon-ion therapy. Phys. Med. Biol. 2017, 63, 01TR02. [Google Scholar] [CrossRef] [PubMed]
- Durante, M. New challenges in high-energy particle radiobiology. Br. J. Radiol. 2014, 87, 20130626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rydberg, B. Clusters of DNA damage induced by ionizing radiation: Formation of short DNA fragments. II. Experimental detection. Radiat. Res. 1996, 145, 200–209. [Google Scholar] [CrossRef] [PubMed]
- Hoglund, E.; Blomquist, E.; Carlsson, J.; Stenerlow, B. DNA damage induced by radiation of different linear energy transfer: Initial fragmentation. Int. J. Radiat. Biol. 2000, 76, 539–547. [Google Scholar] [CrossRef] [PubMed]
- Pang, D.; Chasovskikh, S.; Rodgers, J.E.; Dritschilo, A. Short DNA Fragments Are a Hallmark of Heavy Charged-Particle Irradiation and May Underlie Their Greater Therapeutic Efficacy. Front. Oncol. 2016, 6, 130. [Google Scholar] [CrossRef] [Green Version]
- Konings, K.; Vandevoorde, C.; Baselet, B.; Baatout, S.; Moreels, M. Combination Therapy With Charged Particles and Molecular Targeting: A Promising Avenue to Overcome Radioresistance. Front. Oncol. 2020, 10, 128. [Google Scholar] [CrossRef]
- Held, K.D.; Kawamura, H.; Kaminuma, T.; Paz, A.E.; Yoshida, Y.; Liu, Q.; Willers, H.; Takahashi, A. Effects of Charged Particles on Human Tumor Cells. Front. Oncol. 2016, 6, 23. [Google Scholar] [CrossRef] [Green Version]
- Georgakilas, A.G.; O’Neill, P.; Stewart, R.D. Induction and repair of clustered DNA lesions: What do we know so far? Radiat. Res. 2013, 180, 100–109. [Google Scholar] [CrossRef]
- Durante, M.; Debus, J. Heavy Charged Particles: Does Improved Precision and Higher Biological Effectiveness Translate to Better Outcome in Patients? Semin. Radiat. Oncol. 2018, 28, 160–167. [Google Scholar] [CrossRef]
- Durante, M.; Flanz, J. Charged particle beams to cure cancer: Strengths and challenges. Semin. Oncol. 2019, 46, 219–225. [Google Scholar] [CrossRef] [Green Version]
- Durante, M.; Loeffler, J.S. Charged particles in radiation oncology. Nat. Rev. Clin. Oncol. 2010, 7, 37–43. [Google Scholar] [CrossRef] [PubMed]
- Chiblak, S.; Tang, Z.; Lemke, D.; Knoll, M.; Dokic, I.; Warta, R.; Moustafa, M.; Mier, W.; Brons, S.; Rapp, C.; et al. Carbon irradiation overcomes glioma radioresistance by eradicating stem cells and forming an antiangiogenic and immunopermissive niche. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed]
- Dini, V.; Belli, M.; Tabocchini, M.A. Targeting cancer stem cells: Protons versus photons. Br. J. Radiol. 2020, 93, 20190225. [Google Scholar] [CrossRef] [PubMed]
- Arnold, C.R.; Mangesius, J.; Skvortsova, I.I.; Ganswindt, U. The Role of Cancer Stem Cells in Radiation Resistance. Front. Oncol. 2020, 10, 164. [Google Scholar] [CrossRef] [Green Version]
- Girdhani, S.; Lamont, C.; Hahnfeldt, P.; Abdollahi, A.; Hlatky, L. Proton irradiation suppresses angiogenic genes and impairs cell invasion and tumor growth. Radiat. Res. 2012, 178, 33–45. [Google Scholar] [CrossRef]
- Wozny, A.S.; Vares, G.; Alphonse, G.; Lauret, A.; Monini, C.; Magne, N.; Cuerq, C.; Fujimori, A.; Monboisse, J.C.; Beuve, M.; et al. ROS Production and Distribution: A New Paradigm to Explain the Differential Effects of X-ray and Carbon Ion Irradiation on Cancer Stem Cell Migration and Invasion. Cancers 2019, 11. [Google Scholar] [CrossRef] [Green Version]
- Oonishi, K.; Cui, X.; Hirakawa, H.; Fujimori, A.; Kamijo, T.; Yamada, S.; Yokosuka, O.; Kamada, T. Different effects of carbon ion beams and X-rays on clonogenic survival and DNA repair in human pancreatic cancer stem-like cells. Radiother. Oncol. 2012, 105, 258–265. [Google Scholar] [CrossRef] [Green Version]
- Sai, S.; Wakai, T.; Vares, G.; Yamada, S.; Kamijo, T.; Kamada, T.; Shirai, T. Combination of carbon ion beam and gemcitabine causes irreparable DNA damage and death of radioresistant pancreatic cancer stem-like cells in vitro and in vivo. Oncotarget 2015, 6, 5517–5535. [Google Scholar] [CrossRef] [Green Version]
- Takahashi, A.; Matsumoto, H.; Yuki, K.; Yasumoto, J.; Kajiwara, A.; Aoki, M.; Furusawa, Y.; Ohnishi, K.; Ohnishi, T. High-LET radiation enhanced apoptosis but not necrosis regardless of p53 status. Int. J. Radiat. Oncol. Biol. Phys. 2004, 60, 591–597. [Google Scholar] [CrossRef]
- Yamakawa, N.; Takahashi, A.; Mori, E.; Imai, Y.; Furusawa, Y.; Ohnishi, K.; Kirita, T.; Ohnishi, T. High LET radiation enhances apoptosis in mutated p53 cancer cells through Caspase-9 activation. Cancer Sci. 2008, 99, 1455–1460. [Google Scholar] [CrossRef]
- Mori, E.; Takahashi, A.; Yamakawa, N.; Kirita, T.; Ohnishi, T. High LET heavy ion radiation induces p53-independent apoptosis. J. Radiat. Res. 2009, 50, 37–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ewald, J.A.; Desotelle, J.A.; Wilding, G.; Jarrard, D.F. Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 2010, 102, 1536–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roninson, I.B.; Broude, E.V.; Chang, B.D. If not apoptosis, then what? Treatment-induced senescence and mitotic catastrophe in tumor cells. Drug Resist. Updat. 2001, 4, 303–313. [Google Scholar] [CrossRef] [PubMed]
- Jinno-Oue, A.; Shimizu, N.; Hamada, N.; Wada, S.; Tanaka, A.; Shinagawa, M.; Ohtsuki, T.; Mori, T.; Saha, M.N.; Hoque, A.S.; et al. Irradiation with carbon ion beams induces apoptosis, autophagy, and cellular senescence in a human glioma-derived cell line. Int. J. Radiat. Oncol. Biol. Phys. 2010, 76, 229–241. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Ye, C.; Sun, F.; Wei, W.; Hu, B.; Wang, J. Both Complexity and Location of DNA Damage Contribute to Cellular Senescence Induced by Ionizing Radiation. PLoS ONE 2016, 11, e0155725. [Google Scholar] [CrossRef] [PubMed]
- Wera, A.C.; Lobbens, A.; Stoyanov, M.; Lucas, S.; Michiels, C. Radiation-induced synthetic lethality: Combination of poly(ADP-ribose) polymerase and RAD51 inhibitors to sensitize cells to proton irradiation. Cell Cycle Georget. Tex. 2019, 18, 1770–1783. [Google Scholar] [CrossRef] [PubMed]
- Lecot, P.; Alimirah, F.; Desprez, P.Y.; Campisi, J.; Wiley, C. Context-dependent effects of cellular senescence in cancer development. Br. J. Cancer 2016, 114, 1180–1184. [Google Scholar] [CrossRef] [Green Version]
- Perez-Mancera, P.A.; Young, A.R.; Narita, M. Inside and out: The activities of senescence in cancer. Nat. Rev. 2014, 14, 547–558. [Google Scholar] [CrossRef] [Green Version]
- Yu, Q.; Li, P.; Weng, M.; Wu, S.; Zhang, Y.; Chen, X.; Zhang, Q.; Shen, G.; Ding, X.; Fu, S. Nano-Vesicles are a Potential Tool to Monitor Therapeutic Efficacy of Carbon Ion Radiotherapy in Prostate Cancer. J. Biomed. Nanotechnol. 2018, 14, 168–178. [Google Scholar] [CrossRef]
- Fong, P.C.; Boss, D.S.; Yap, T.A.; Tutt, A.; Wu, P.; Mergui-Roelvink, M.; Mortimer, P.; Swaisland, H.; Lau, A.; O’Connor, M.J.; et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N. Engl. J. Med. 2009, 361, 123–134. [Google Scholar] [CrossRef] [Green Version]
- Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef] [PubMed]
- Szymonowicz, K.; Krysztofiak, A.; Linden, J.V.; Kern, A.; Deycmar, S.; Oeck, S.; Squire, A.; Koska, B.; Hlouschek, J.; Vullings, M.; et al. Proton Irradiation Increases the Necessity for Homologous Recombination Repair Along with the Indispensability of Non-Homologous End Joining. Cells 2020, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fontana, A.O.; Augsburger, M.A.; Grosse, N.; Guckenberger, M.; Lomax, A.J.; Sartori, A.A.; Pruschy, M.N. Differential DNA repair pathway choice in cancer cells after proton- and photon-irradiation. Radiother. Oncol. 2015, 116, 374–380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ward, A.; Khanna, K.K.; Wiegmans, A.P. Targeting homologous recombination, new pre-clinical and clinical therapeutic combinations inhibiting RAD51. Cancer Treat. Rev. 2015, 41, 35–45. [Google Scholar] [CrossRef]
- Maacke, H.; Jost, K.; Opitz, S.; Miska, S.; Yuan, Y.; Hasselbach, L.; Lüttges, J.; Kalthoff, H.; Stürzbecher, H.W. DNA repair and recombination factor Rad51 is over-expressed in human pancreatic adenocarcinoma. Oncogene 2000, 19, 2791–2795. [Google Scholar] [CrossRef]
- Han, H.; Bearss, D.J.; Browne, L.W.; Calaluce, R.; Nagle, R.B.; Von Hoff, D.D. Identification of Differentially Expressed Genes in Pancreatic Cancer Cells Using cDNA Microarray. Cancer Res. 2002, 62, 2890–2896. [Google Scholar]
- Alagpulinsa, D.A.; Ayyadevara, S.; Shmookler Reis, R.J. A Small-Molecule Inhibitor of RAD51 Reduces Homologous Recombination and Sensitizes Multiple Myeloma Cells to Doxorubicin. Front. Oncol. 2014, 4, 289. [Google Scholar] [CrossRef] [Green Version]
- Huang, F.; Mazina, O.M.; Zentner, I.J.; Cocklin, S.; Mazin, A.V. Inhibition of homologous recombination in human cells by targeting RAD51 recombinase. J. Med. Chem. 2012, 55, 3011–3020. [Google Scholar] [CrossRef]
- Ma, H.; Takahashi, A.; Yoshida, Y.; Adachi, A.; Kanai, T.; Ohno, T.; Nakano, T. Combining carbon ion irradiation and non-homologous end-joining repair inhibitor NU7026 efficiently kills cancer cells. Radiat. Oncol. 2015, 10, 225. [Google Scholar] [CrossRef] [Green Version]
- Huang, F.; Mazin, A.V. A small molecule inhibitor of human RAD51 potentiates breast cancer cell killing by therapeutic agents in mouse xenografts. PLoS ONE 2014, 9, e100993. [Google Scholar] [CrossRef]
- Huang, F.; Motlekar, N.A.; Burgwin, C.M.; Napper, A.D.; Diamond, S.L.; Mazin, A.V. Identification of specific inhibitors of human RAD51 recombinase using high-throughput screening. ACS Chem. Biol. 2011, 6, 628–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Wang, X.; Zhang, P.; Wang, Y. The Ku-dependent non-homologous end-joining but not other repair pathway is inhibited by high linear energy transfer ionizing radiation. DNA Repair 2008, 7, 725–733. [Google Scholar] [CrossRef] [PubMed]
- Iliakis, G.; Murmann, T.; Soni, A. Alternative end-joining repair pathways are the ultimate backup for abrogated classical non-homologous end-joining and homologous recombination repair: Implications for the formation of chromosome translocations. Mutat. Res. Genet. Toxicol. Environ. Mutagenesis 2015, 793, 166–175. [Google Scholar] [CrossRef] [PubMed]
- Chan, N.; Pires, I.M.; Bencokova, Z.; Coackley, C.; Luoto, K.R.; Bhogal, N.; Lakshman, M.; Gottipati, P.; Oliver, F.J.; Helleday, T.; et al. Contextual synthetic lethality of cancer cell kill based on the tumor microenvironment. Cancer Res. 2010, 70, 8045–8054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, N.; Koritzinsky, M.; Zhao, H.; Bindra, R.; Glazer, P.M.; Powell, S.; Belmaaza, A.; Wouters, B.; Bristow, R.G. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance. Cancer Res. 2008, 68, 605–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bagnolini, G.; Milano, D.; Manerba, M.; Schipani, F.; Ortega, J.A.; Gioia, D.; Falchi, F.; Balboni, A.; Farabegoli, F.; De Franco, F.; et al. Synthetic Lethality in Pancreatic Cancer: Discovery of a New RAD51-BRCA2 Small Molecule Disruptor That Inhibits Homologous Recombination and Synergizes with Olaparib. J. Med. Chem. 2020, 63, 2588–2619. [Google Scholar] [CrossRef]
- Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J. Immunother. 2010, 33, 828–833. [Google Scholar] [CrossRef]
- Henriksen, A.; Dyhl-Polk, A.; Chen, I.; Nielsen, D. Checkpoint inhibitors in pancreatic cancer. Cancer Treat. Rev. 2019, 78, 17–30. [Google Scholar] [CrossRef]
- Fridman, W.H.; Zitvogel, L.; Sautes-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
- Lankadasari, M.B.; Mukhopadhyay, P.; Mohammed, S.; Harikumar, K.B. TAMing pancreatic cancer: Combat with a double edged sword. Mol. Cancer 2019, 18, 48. [Google Scholar] [CrossRef] [Green Version]
- Hu, H.; Hang, J.J.; Han, T.; Zhuo, M.; Jiao, F.; Wang, L.W. The M2 phenotype of tumor-associated macrophages in the stroma confers a poor prognosis in pancreatic cancer. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2016, 37, 8657–8664. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Herndon, J.M.; Sojka, D.K.; Kim, K.W.; Knolhoff, B.L.; Zuo, C.; Cullinan, D.R.; Luo, J.; Bearden, A.R.; Lavine, K.J.; et al. Tissue-Resident Macrophages in Pancreatic Ductal Adenocarcinoma Originate from Embryonic Hematopoiesis and Promote Tumor Progression. Immunity 2017, 47, 323–338.e6. [Google Scholar] [CrossRef]
- Germano, G.; Frapolli, R.; Belgiovine, C.; Anselmo, A.; Pesce, S.; Liguori, M.; Erba, E.; Uboldi, S.; Zucchetti, M.; Pasqualini, F.; et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 2013, 23, 249–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borgoni, S.; Iannello, A.; Cutrupi, S.; Allavena, P.; D’Incalci, M.; Novelli, F.; Cappello, P. Depletion of tumor-associated macrophages switches the epigenetic profile of pancreatic cancer infiltrating T cells and restores their anti-tumor phenotype. Oncoimmunology 2018, 7, e1393596. [Google Scholar] [CrossRef] [PubMed]
- Guo, Z.; Wang, H.; Meng, F.; Li, J.; Zhang, S. Combined Trabectedin and anti-PD1 antibody produces a synergistic antitumor effect in a murine model of ovarian cancer. J. Transl. Med. 2015, 13, 247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Mayer, A.; Deshpande, A.D.; Carpenter, D.; Mitchem, J.B.; Plambeck-Suess, S.M.; Worley, L.A.; Goetz, B.D.; et al. Inflammatory monocyte mobilization decreases patient survival in pancreatic cancer: A role for targeting the CCL2/CCR2 axis. Clin. Cancer Res. 2013, 19, 3404–3415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nywening, T.M.; Wang-Gillam, A.; Sanford, D.E.; Belt, B.A.; Panni, R.Z.; Cusworth, B.M.; Toriola, A.T.; Nieman, R.K.; Worley, L.A.; Yano, M.; et al. Targeting tumour-associated macrophages with CCR2 inhibition in combination with FOLFIRINOX in patients with borderline resectable and locally advanced pancreatic cancer: A single-centre, open-label, dose-finding, non-randomised, phase 1b trial. Lancet Oncol. 2016, 17, 651–662. [Google Scholar] [CrossRef] [Green Version]
- Candido, J.B.; Morton, J.P.; Bailey, P.; Campbell, A.D.; Karim, S.A.; Jamieson, T.; Lapienyte, L.; Gopinathan, A.; Clark, W.; McGhee, E.J.; et al. CSF1R(+) Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype. Cell Rep. 2018, 23, 1448–1460. [Google Scholar] [CrossRef] [Green Version]
- Mitchem, J.B.; Brennan, D.J.; Knolhoff, B.L.; Belt, B.A.; Zhu, Y.; Sanford, D.E.; Belaygorod, L.; Carpenter, D.; Collins, L.; Piwnica-Worms, D.; et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013, 73, 1128–1141. [Google Scholar] [CrossRef]
- Seifert, L.; Werba, G.; Tiwari, S.; Giao Ly, N.N.; Nguy, S.; Alothman, S.; Alqunaibit, D.; Avanzi, A.; Daley, D.; Barilla, R.; et al. Radiation Therapy Induces Macrophages to Suppress T-Cell Responses Against Pancreatic Tumors in Mice. Gastroenterology 2016, 150, 1659–1672. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Knolhoff, B.L.; Meyer, M.A.; Nywening, T.M.; West, B.L.; Luo, J.; Wang-Gillam, A.; Goedegebuure, S.P.; Linehan, D.C.; DeNardo, D.G. CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res. 2014, 74, 5057–5069. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; Chiorean, E.G.; Fishman, M.P.; Saboury, B.; Teitelbaum, U.R.; Sun, W.; Huhn, R.D.; Song, W.; Li, D.; Sharp, L.L.; et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011, 331, 1612–1616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cullis, J.; Siolas, D.; Avanzi, A.; Barui, S.; Maitra, A.; Bar-Sagi, D. Macropinocytosis of Nab-paclitaxel Drives Macrophage Activation in Pancreatic Cancer. Cancer Immunol. Res. 2017, 5, 182–190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wanderley, C.W.; Colon, D.F.; Luiz, J.P.M.; Oliveira, F.F.; Viacava, P.R.; Leite, C.A.; Pereira, J.A.; Silva, C.M.; Silva, C.R.; Silva, R.L.; et al. Paclitaxel Reduces Tumor Growth by Reprogramming Tumor-Associated Macrophages to an M1 Profile in a TLR4-Dependent Manner. Cancer Res. 2018, 78, 5891–5900. [Google Scholar] [CrossRef] [Green Version]
- Prakash, H.; Nadella, V.; Singh, S.; Schmitz-Winnenthal, H. CD14/TLR4 priming potentially recalibrates and exerts anti-tumor efficacy in tumor associated macrophages in a mouse model of pancreatic carcinoma. Sci. Rep. 2016, 6, 31490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carvalho, H.A.; Villar, R.C. Radiotherapy and immune response: The systemic effects of a local treatment. Clinics 2018, 73, e557s. [Google Scholar] [CrossRef]
- Arina, A.; Gutiontov, S.I.; Weichselbaum, R.R. Radiotherapy and Immunotherapy for Cancer: From “Systemic” to “Multisite”. Clin. Cancer Res. 2020. [Google Scholar] [CrossRef]
- Burnette, B.; Weichselbaum, R.R. Radiation as an immune modulator. Semin. Radiat. Oncol. 2013, 23, 273–280. [Google Scholar] [CrossRef]
- Azad, A.; Yin Lim, S.; D’Costa, Z.; Jones, K.; Diana, A.; Sansom, O.J.; Kruger, P.; Liu, S.; McKenna, W.G.; Dushek, O.; et al. PD-L1 blockade enhances response of pancreatic ductal adenocarcinoma to radiotherapy. EMBO Mol. Med. 2017, 9, 167–180. [Google Scholar] [CrossRef]
- Yamamoto, K.; Venida, A.; Yano, J.; Biancur, D.E.; Kakiuchi, M.; Gupta, S.; Sohn, A.S.W.; Mukhopadhyay, S.; Lin, E.Y.; Parker, S.J.; et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 2020. [Google Scholar] [CrossRef]
- Wolpin, B.M.; Rubinson, D.A.; Wang, X.; Chan, J.A.; Cleary, J.M.; Enzinger, P.C.; Fuchs, C.S.; McCleary, N.J.; Meyerhardt, J.A.; Ng, K.; et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. Oncology 2014, 19, 637–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- New, M.; Tooze, S. The Role of Autophagy in Pancreatic Cancer-Recent Advances. Biology 2019, 9, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef] [PubMed]
- Kalbasi, A.; Komar, C.; Tooker, G.M.; Liu, M.; Lee, J.W.; Gladney, W.L.; Ben-Josef, E.; Beatty, G.L. Tumor-Derived CCL2 Mediates Resistance to Radiotherapy in Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2017, 23, 137–148. [Google Scholar] [CrossRef] [Green Version]
- Klug, F.; Prakash, H.; Huber, P.E.; Seibel, T.; Bender, N.; Halama, N.; Pfirschke, C.; Voss, R.H.; Timke, C.; Umansky, L.; et al. Low-dose irradiation programs macrophage differentiation to an iNOS(+)/M1 phenotype that orchestrates effective T cell immunotherapy. Cancer Cell 2013, 24, 589–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prakash, H.; Klug, F.; Nadella, V.; Mazumdar, V.; Schmitz-Winnenthal, H.; Umansky, L. Low doses of gamma irradiation potentially modifies immunosuppressive tumor microenvironment by retuning tumor-associated macrophages: Lesson from insulinoma. Carcinogenesis 2016, 37, 301–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Genard, G.; Lucas, S.; Michiels, C. Reprogramming of Tumor-Associated Macrophages with Anticancer Therapies: Radiotherapy versus Chemo- and Immunotherapies. Front. Immunol. 2017, 8, 828. [Google Scholar] [CrossRef] [Green Version]
- Durante, M.; Formenti, S. Harnessing radiation to improve immunotherapy: Better with particles? Br. J. Radiol. 2020, 93, 20190224. [Google Scholar] [CrossRef]
- Gameiro, S.R.; Jammeh, M.L.; Wattenberg, M.M.; Tsang, K.Y.; Ferrone, S.; Hodge, J.W. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 2014, 5, 403–416. [Google Scholar] [CrossRef] [Green Version]
- Gameiro, S.R.; Malamas, A.S.; Bernstein, M.B.; Tsang, K.Y.; Vassantachart, A.; Sahoo, N.; Tailor, R.; Pidikiti, R.; Guha, C.P.; Hahn, S.M.; et al. Tumor Cells Surviving Exposure to Proton or Photon Radiation Share a Common Immunogenic Modulation Signature, Rendering Them More Sensitive to T Cell-Mediated Killing. Int. J. Radiat. Oncol. Biol. Phys. 2016, 95, 120–130. [Google Scholar] [CrossRef] [Green Version]
- Jin, X.; Li, F.; Zheng, X.; Liu, Y.; Hirayama, R.; Liu, X.; Li, P.; Zhao, T.; Dai, Z.; Li, Q. Carbon ions induce autophagy effectively through stimulating the unfolded protein response and subsequent inhibiting Akt phosphorylation in tumor cells. Sci. Rep. 2015, 5, 13815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, X.; Liu, Y.; Ye, F.; Liu, X.; Furusawa, Y.; Wu, Q.; Li, F.; Zheng, X.; Dai, Z.; Li, Q. Role of autophagy in high linear energy transfer radiation-induced cytotoxicity to tumor cells. Cancer Sci. 2014, 105, 770–778. [Google Scholar] [CrossRef] [PubMed]
- Genard, G.; Wera, A.C.; Huart, C.; Le Calve, B.; Penninckx, S.; Fattaccioli, A.; Tabarrant, T.; Demazy, C.; Ninane, N.; Heuskin, A.C.; et al. Proton irradiation orchestrates macrophage reprogramming through NFkappaB signaling. Cell Death Dis. 2018, 9, 728. [Google Scholar] [CrossRef] [PubMed]
- Buonanno, M.; Grilj, V.; Brenner, D.J. Biological effects in normal cells exposed to FLASH dose rate protons. Radiother. Oncol. 2019, 139, 51–55. [Google Scholar] [CrossRef] [PubMed]
- Girdhani, S.; Abel, E.; Katsis, A.; Rodriquez, A.; Senapati, S.; KuVillanueva, A.; Jackson, I.L.; Eley, J.; Vujaskovic, Z.; Parry, R. Abstract LB-280: FLASH: A novel paradigm changing tumor irradiation platform that enhances therapeutic ratio by reducing normal tissue toxicity and activating immune pathways. Cancer Res. 2019, 79, LB-280. [Google Scholar] [CrossRef]
- Rama, N.; Saha, T.; Shukla, S.; Goda, C.; Milewski, D.; Mascia, A.E.; Vatner, R.E.; Sengupta, D.; Katsis, A.; Abel, E.; et al. Improved Tumor Control Through T-cell Infiltration Modulated by Ultra-High Dose Rate Proton FLASH Using a Clinical Pencil Beam Scanning Proton System. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, S164–S165. [Google Scholar] [CrossRef]
- Dutt, S.; Ahmed, M.M.; Loo, B.W., Jr.; Strober, S. Novel Radiation Therapy Paradigms and Immunomodulation: Heresies and Hope. Semin. Radiat. Oncol. 2020, 30, 194–200. [Google Scholar] [CrossRef]
- Sato, H.; Okonogi, N.; Nakano, T. Rationale of combination of anti-PD-1/PD-L1 antibody therapy and radiotherapy for cancer treatment. Int. J. Clin. Oncol. 2020, 25, 801–809. [Google Scholar] [CrossRef] [Green Version]
- Yelamos, J.; Moreno-Lama, L.; Jimeno, J.; Ali, S.O. Immunomodulatory Roles of PARP-1 and PARP-2: Impact on PARP-Centered Cancer Therapies. Cancers 2020, 12, 392. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharya, S.; Srinivasan, K.; Abdisalaam, S.; Su, F.; Raj, P.; Dozmorov, I.; Mishra, R.; Wakeland, E.K.; Ghose, S.; Mukherjee, S.; et al. RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res. 2017, 45, 4590–4605. [Google Scholar] [CrossRef] [Green Version]
ID | Condition | Year | Status | N | Radiation | Dose | Concurrent Chemotherapy |
---|---|---|---|---|---|---|---|
NCT03885284 | R-PDAC (adjuvant) | 2019 | Recruiting | 12 | Protons | 25 Gy (RBE) in five fractions | mFOLFIRINOX |
NCT01591733 | R-PDAC (neoadjuvant) | 2012 | Active | 48 | Protons (Photons) | 25 Gy (RBE) in five fractions (30 Gy in 10 fractions) | FOLFIRINOX + Capecitabine |
NCT01494155 | R-PDAC (neoadjvant) | 2011 | Active | 50 | Protons (Photons) | 25 Gy (RBE) in five fractions (30 Gy in 10 fractions) | Capecitabine + Hydroxychloroquine |
NCT03822936 | R-PDAC (neoadjuvant) | 2019 | Recruiting | 30 | Carbon ions | 38.4 Gy (RBE) in eight fractions | N/A |
NCT02598349 | Unresectable | 2015 | Recruiting | 60 | Protons | 63 Gy (RBE) in 28 fractions | Capecitabine |
NCT04194268 | Unresectable | 2019 | Recruiting | 25 | Carbon ions | 48 Gy (RBE) in 12 fractions | N/A |
NCT03652428 | LA-PDAC | 2018 | Recruiting | 24 | Protons | 75 Gy (RBE) in 15 fractions | Gemcitabine |
NCT03652428 | LA-PDAC | 2018 | Recruiting | 24 | Protons | 75 Gy (RBE)E in 15 fractions | Nab-paclitaxel + Gemcitabine |
NCT04082455 | LA-PDAC | 2019 | Recruiting | 49 | Carbon ions | 60-67.5Gy (RBE) in 15 fractions | N/A |
NCT01821729 | LA-PDAC | 2013 | Active | 50 | Protons (Photons) | 25 Gy (RBE) in five fractions (if persistent vascular involvement 50.4 Gy with vascular boost to 58.8 Gy) | FOLFIRIRINOX + Losartan |
NCT03536182 | LA-PDAC | 2018 | Active | 110 | Carbon ions (Photons) | 55.2 or 57.6 Gy (RBE) in 12 fractions (50.4–56 Gy in 28 fractions) | • Gemcitabine • Gemcitabine + Nab-paclitaxel • FOLFIRINOX |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Huart, C.; Chen, J.-W.; Le Calvé, B.; Michiels, C.; Wéra, A.-C. Could Protons and Carbon Ions Be the Silver Bullets Against Pancreatic Cancer? Int. J. Mol. Sci. 2020, 21, 4767. https://doi.org/10.3390/ijms21134767
Huart C, Chen J-W, Le Calvé B, Michiels C, Wéra A-C. Could Protons and Carbon Ions Be the Silver Bullets Against Pancreatic Cancer? International Journal of Molecular Sciences. 2020; 21(13):4767. https://doi.org/10.3390/ijms21134767
Chicago/Turabian StyleHuart, Camille, Jia-Wei Chen, Benjamin Le Calvé, Carine Michiels, and Anne-Catherine Wéra. 2020. "Could Protons and Carbon Ions Be the Silver Bullets Against Pancreatic Cancer?" International Journal of Molecular Sciences 21, no. 13: 4767. https://doi.org/10.3390/ijms21134767
APA StyleHuart, C., Chen, J. -W., Le Calvé, B., Michiels, C., & Wéra, A. -C. (2020). Could Protons and Carbon Ions Be the Silver Bullets Against Pancreatic Cancer? International Journal of Molecular Sciences, 21(13), 4767. https://doi.org/10.3390/ijms21134767