The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors
Abstract
:1. Introduction
2. ICI for Primary Brain Tumors
2.1. Glioblastoma
2.2. Isocitrate Dehydrogenase (IDH) Mutant Gliomas
Author | Phase | Regimen | Treatment |
---|---|---|---|
NCT03557359 | II | Anti PD-1 monotherapy | Nivolumab |
NCT03893903 | I | IDH vax + anti-PD-L1 | IDH-1 vaccine + Avelumab |
NCT04056910 | II | Anti-PD-1 + IDH1-inhibitor | Nivolumab + Ivosidenib |
NCT02968940 | II | Anti-PDL1 + radiation | Avelumab + HFRT |
2.3. Meningioma
2.4. Primary Central Nervous System Lymphoma
3. ICI for Brain Metastases
3.1. Parenchymal Metastases
3.2. Melanoma
3.3. Breast Cancer
3.4. Non-Small Cell Lung Cancer
3.5. Leptomeningeal Metastases
4. Discussion
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Wei, S.C.; Duffy, C.R.; Allison, J.P. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov. 2018, 8, 1069–1086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Twomey, J.D.; Zhang, B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J. 2021, 23, 39. [Google Scholar] [CrossRef]
- Rossi, E.; Schinzari, G.; Maiorano, B.A.; Indellicati, G.; Di Stefani, A.; Pagliara, M.M.; Fragomeni, S.M.; De Luca, E.V.; Sammarco, M.G.; Garganese, G.; et al. Efficacy of immune checkpoint inhibitors in different types of melanoma. Hum. Vaccin. Immunother. 2021, 17, 4–13. [Google Scholar] [CrossRef]
- Berghmans, T.; Durieux, V.; Hendriks, L.E.L.; Dingemans, A.M. Immunotherapy: From Advanced NSCLC to Early Stages, an Evolving Concept. Front. Med. 2020, 7, 90. [Google Scholar] [CrossRef] [Green Version]
- Tomaszewski, W.; Sanchez-Perez, L.; Gajewski, T.F.; Sampson, J.H. Brain Tumor Microenvironment and Host State: Implications for Immunotherapy. Clin. Cancer Res. 2019, 25, 4202–4210. [Google Scholar] [CrossRef] [Green Version]
- Medikonda, R.; Dunn, G.; Rahman, M.; Fecci, P.; Lim, M. A review of glioblastoma immunotherapy. J. Neuro-Oncol. 2021, 151, 41–53. [Google Scholar] [CrossRef]
- Sampson, J.H.; Gunn, M.D.; Fecci, P.E.; Ashley, D.M. Brain immunology and immunotherapy in brain tumours. Nat. Rev. Cancer 2020, 20, 12–25. [Google Scholar] [CrossRef] [PubMed]
- Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stupp, R.; Hegi, M.E.; Mason, W.P.; Van Den Bent, M.J.; Taphoorn, M.J.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef] [PubMed]
- Lamborn, K.R.; Yung, W.K.A.; Chang, S.M.; Wen, P.Y.; Cloughesy, T.F.; DeAngelis, L.; Robins, H.I.; Lieberman, F.S.; Fine, H.A.; Fink, K.L.; et al. Progression-free survival: An important end point in evaluating therapy for recurrent high-grade gliomas. Neuro-Oncol. 2008, 10, 162–170. [Google Scholar] [CrossRef]
- Wu, W.; Lamborn, K.R.; Buckner, J.C.; Novotny, P.J.; Chang, S.M.; O’Fallon, J.R.; Jaeckle, K.A.; Prados, M.D. Joint NCCTG and NABTC prognostic factors analysis for high-grade recurrent glioma. Neuro-Oncol. 2009, 12, 164–172. [Google Scholar] [CrossRef] [Green Version]
- Clarke, J.L.; Ennis, M.M.; Yung, W.K.A.; Chang, S.M.; Wen, P.Y.; Cloughesy, T.F.; DeAngelis, L.; Robins, H.I.; Lieberman, F.S.; Fine, H.A.; et al. Is surgery at progression a prognostic marker for improved 6-month progression-free survival or overall survival for patients with recurrent glioblastoma? Neuro-Oncol. 2011, 13, 1118–1124. [Google Scholar] [CrossRef]
- Reardon, D.A.; Kim, T.M.; Frenel, J.; Simonelli, M.; Lopez, J.; Subramaniam, D.S.; Siu, L.L.; Wang, H.; Krishnan, S.; Stein, K.; et al. Treatment with pembrolizumab in programmed death ligand 1–positive recurrent glioblastoma: Results from the multicohort phase 1 KEYNOTE-028 trial. Cancer 2021, 127, 1620–1629. [Google Scholar] [CrossRef] [PubMed]
- Berghoff, A.S.; Kiesel, B.; Widhalm, G.; Rajky, O.; Ricken, G.; Wöhrer, A.; Dieckmann, K.; Filipits, M.; Brandstetter, A.; Weller, M.; et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro-Oncol. 2015, 17, 1064–1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilmotte, R.; Burkhardt, K.; Kindler, V.; Belkouch, M.-C.; Dussex, G.; De Tribolet, N.; Walker, P.R.; Dietrich, P.-Y. B7-homolog 1 expression by human glioma: A new mechanism of immune evasion. Neuroreport 2005, 16, 1081–1085. [Google Scholar] [CrossRef] [PubMed]
- Xue, S.; Song, G.; Yu, J. The prognostic significance of PD-L1 expression in patients with glioma: A meta-analysis. Sci. Rep. 2017, 7, 4231. [Google Scholar] [CrossRef] [Green Version]
- Nayak, L.; Molinaro, A.M.; Peters, K.B.; Clarke, J.L.; Jordan, J.T.; de Groot, J.F.; Nghiemphu, P.L.; Kaley, T.J.; Colman, H.; McCluskey, C.; et al. Randomized Phase II and Biomarker Study of Pembrolizumab plus Bevacizumab versus Pembrolizumab Alone for Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2021, 27, 1048–1057. [Google Scholar] [CrossRef]
- Nayak, L.; Standifer, N.; Dietrich, J.; Clarke, J.L.; Dunn, G.P.; Lim, M.; Cloughesy, T.; Gan, H.K.; Flagg, E.; George, E.; et al. Circulating Immune Cell and Outcome Analysis from the Phase II Study of PD-L1 Blockade with Durvalumab for Newly Diagnosed and Recurrent Glioblastoma. Clin. Cancer Res. 2022, 28, 2567–2578. [Google Scholar] [CrossRef]
- Lukas, R.V.; Rodon, J.; Becker, K.; Wong, E.T.; Shih, K.; Touat, M.; Fassò, M.; Osborne, S.; Molinero, L.; O’Hear, C.; et al. Clinical activity and safety of atezolizumab in patients with recurrent glioblastoma. J. Neuro-Oncol. 2018, 140, 317–328. [Google Scholar] [CrossRef]
- Chiocca, E.A.; Gelb, A.B.; Chen, C.C.; Rao, G.; A Reardon, D.; Wen, P.Y.; Bi, W.L.; Peruzzi, P.; Amidei, C.; Triggs, D.; et al. Combined immunotherapy with controlled interleukin-12 gene therapy and immune checkpoint blockade in recurrent glioblastoma: An open-label, multi-institutional phase I trial. Neuro-Oncol. 2021, 24, 951–963. [Google Scholar] [CrossRef]
- Omuro, A.; A Reardon, D.; Sampson, J.H.; Baehring, J.; Sahebjam, S.; Cloughesy, T.F.; Chalamandaris, A.-G.; Von Potter, V.; Butowski, N.; Lim, M. Nivolumab plus radiotherapy with or without temozolomide in newly diagnosed glioblastoma: Results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncol. Adv. 2022, 4, vdac025. [Google Scholar] [CrossRef]
- Omuro, A.; Vlahovic, G.; Lim, M.; Sahebjam, S.; Baehring, J.; Cloughesy, T.; Voloschin, A.; Ramkissoon, S.H.; Ligon, K.L.; Latek, R.; et al. Nivolumab with or without ipilimumab in patients with recurrent glioblastoma: Results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncol. 2018, 20, 674–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schalper, K.A.; Rodriguez-Ruiz, M.E.; Diez-Valle, R.; López-Janeiro, A.; Porciuncula, A.; Idoate, M.A.; Inogés, S.; De Andrea, C.; De Cerio, A.L.-D.; Tejada, S.; et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 2019, 25, 470–476. [Google Scholar] [CrossRef]
- Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R.; Hugo, W.; Lee, A.H.; Davidson, T.B.; Wang, A.C.; Ellingson, B.M.; Rytlewski, J.A.; Sanders, C.M.; et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef]
- Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial Supplemental content. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
- Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro-Oncol. 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
- Arjaans, M.; Oosting, S.F.; Schröder, C.P.; de Vries, E.G. Bevacizumab-Induced Vessel Normalization Hampers Tumor Uptake of Antibodies—Response. Cancer Res. 2013, 73, 7147–7148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, A.H.; Sun, L.; Mochizuki, A.Y.; Reynoso, J.G.; Orpilla, J.; Chow, F.; Kienzler, J.C.; Everson, R.G.; Nathanson, D.A.; Bensinger, S.J.; et al. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma. Nat. Commun. 2021, 12, 6938. [Google Scholar] [CrossRef]
- Omuro, A.; A Brandes, A.; Carpentier, A.F.; Idbaih, A.; A Reardon, D.; Cloughesy, T.; Sumrall, A.; Baehring, J.; Bent, M.V.D.; Bähr, O.; et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro-Oncol. 2022, 25, 123–134. [Google Scholar] [CrossRef] [PubMed]
- Nduom, E.K.; Wei, J.; Yaghi, N.K.; Huang, N.; Kong, L.-Y.; Gabrusiewicz, K.; Ling, X.; Zhou, S.; Ivan, C.; Chen, J.Q.; et al. PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncol. 2016, 18, 195–205. [Google Scholar] [CrossRef] [Green Version]
- Wang, P.; Fang, X.; Yin, T.; Tian, H.; Yu, J.; Teng, F. Efficacy and Safety of Anti-PD-1 Plus Anlotinib in Patients With Advanced Non–Small-Cell Lung Cancer After Previous Systemic Treatment Failure—A Retrospective Study. Front. Oncol. 2021, 11, 628124. [Google Scholar] [CrossRef]
- Klemm, F.; Maas, R.R.; Bowman, R.L.; Kornete, M.; Soukup, K.; Nassiri, S.; Brouland, J.-P.; Iacobuzio-Donahue, C.A.; Brennan, C.; Tabar, V.; et al. Interrogation of the Microenvironmental Landscape in Brain Tumors Reveals Disease-Specific Alterations of Immune Cells. Cell 2020, 181, 1643–1660.e17. [Google Scholar] [CrossRef]
- Phillips, J.P.; Eremin, O.; Anderson, J.R. Lymphoreticuar cells in human brain tumors and in the normal brain. Br. J. Cancer. 1982, 45, 61–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Darmanis, S.; Sloan, S.A.; Croote, D.; Mignardi, M.; Chernikova, S.; Samghababi, P.; Zhang, Y.; Neff, N.; Kowarsky, M.; Caneda, C.; et al. Single-Cell RNA-Seq Analysis of Infiltrating Neoplastic Cells at the Migrating Front of Human Glioblastoma. Cell Rep. 2017, 21, 1399–1410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gieryng, A.; Pszczolkowska, D.; Walentynowicz, K.A.; Rajan, W.D.; Kaminska, B. Immune microenvironment of gliomas. Lab. Investig. 2017, 97, 498–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venteicher, A.S.; Tirosh, I.; Hebert, C.; Yizhak, K.; Neftel, C.; Filbin, M.G.; Hovestadt, V.; Escalante, L.E.; Shaw, M.L.; Rodman, C.; et al. Decoupling genetics, lineages, and microenvironment in IDH-mutant gliomas by single-cell RNA-seq. Science 2017, 355, eaai8478. [Google Scholar] [CrossRef] [Green Version]
- Andersen, B.M.; Akl, C.F.; Wheeler, M.A.; Chiocca, E.A.; Reardon, D.A.; Quintana, F.J. Glial and myeloid heterogeneity in the brain tumour microenvironment. Nat. Rev. Cancer 2021, 21, 786–802. [Google Scholar] [CrossRef]
- Miller, J.J.; Gonzalez Castro, L.N.; McBrayer, S.; Weller, M.; Cloughesy, T.; Portnow, J.; Andronesi, O.; Barnholtz-Sloan, J.S.; Baumert, B.G.; Berger, M.S.; et al. Isocitrate dehydrogenase (IDH) mutant gliomas: A Society for Neuro-Oncology (SNO) consensus review on diagnosis, management, and future directions. Neuro-Oncol. 2022, 25, 4–25. [Google Scholar] [CrossRef] [PubMed]
- Bunse, L.; Pusch, S.; Bunse, T.; Sahm, F.; Sanghvi, K.; Friedrich, M.; Alansary, D.; Sonner, J.K.; Green, E.; Deumelandt, K.; et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 2018, 24, 1192–1203. [Google Scholar] [CrossRef]
- Wang, Z.; Zhang, C.; Liu, X.; Wang, Z.; Sun, L.; Li, G.; Liang, J.; Hu, H.; Liu, Y.; Zhang, W.; et al. Molecular and clinical characterization of PD-L1 expression at transcriptional level via 976 samples of brain glioma. Oncoimmunology 2016, 5, e1196310. [Google Scholar] [CrossRef] [Green Version]
- Mu, L.; Long, Y.; Yang, C.; Jin, L.; Tao, H.; Ge, H.; Chang, Y.E.; Karachi, A.; Kubilis, P.S.; De Leon, G.; et al. The IDH1 Mutation-Induced Oncometabolite, 2-Hydroxyglutarate, May Affect DNA Methylation and Expression of PD-L1 in Gliomas. Front. Mol. Neurosci. 2018, 11, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Röver, L.K.; Gevensleben, H.; Dietrich, J.; Bootz, F.; Landsberg, J.; Goltz, D.; Dietrich, D. PD-1 (PDCD1) Promoter Methylation Is a Prognostic Factor in Patients With Diffuse Lower-Grade Gliomas Harboring Isocitrate Dehydrogenase (IDH) Mutations. Ebiomedicine 2018, 28, 97–104. [Google Scholar] [CrossRef] [Green Version]
- Mellinghoff, I.K.; Ellingson, B.M.; Touat, M.; Maher, E.; Macarena, I.; Holdhoff, M.; Cote, G.M.; Burris, H.; Janku, F.; Young, R.J.; et al. Ivosidenib in Isocitrate Dehydrogenase 1-Mutated Advanced Glioma. J. Clin. Oncol. 2020, 38, 3398. [Google Scholar] [CrossRef]
- Natsume, A.; Wakabayashi, T.; Miyakita, Y.; Narita, Y.; Mineharu, Y.; Arakawa, Y.; Yamasaki, F.; Sugiyama, K.; Hata, N.; Muragaki, Y.; et al. Phase I study of a brain penetrant mutant IDH1 inhibitor DS-1001b in patients with recurrent or progressive IDH1 mutant gliomas. J. Clin. Oncol. 2019, 37, 2004. [Google Scholar] [CrossRef]
- Bi, W.L.; Nayak, L.; Meredith, D.M.; Driver, J.; Du, Z.; Hoffman, S.; Li, Y.; Lee, E.Q.; Beroukhim, R.; Rinne, M.; et al. Activity of PD-1 blockade with nivolumab among patients with recurrent atypical/anaplastic meningioma: Phase II trial results. Neuro-Oncol. 2022, 24, 101–113. [Google Scholar] [CrossRef] [PubMed]
- Brastianos, P.K.; Kim, A.E.; Giobbie-Hurder, A.; Lee, E.Q.; Wang, N.; Eichler, A.F.; Chukwueke, U.; Forst, D.A.; Arrillaga-Romany, I.C.; Dietrich, J.; et al. Phase 2 study of pembrolizumab in patients with recurrent and residual high-grade meningiomas. Nat. Commun. 2022, 13, 1325. [Google Scholar] [CrossRef]
- Nidamanuri, P.; Drappatz, J. Immune checkpoint inhibitor therapy for recurrent meningiomas: A retrospective chart review. J. Neuro-Oncol. 2022, 157, 271–276. [Google Scholar] [CrossRef]
- Kaley, T.; Barani, I.; Chamberlain, M.; McDermott, M.; Panageas, K.; Raizer, J.; Rogers, L.; Schiff, D.; Vogelbaum, M.; Weber, D.; et al. Historical benchmarks for medical therapy trials in surgery- and radiation-refractory meningioma: A RANO review. Neuro-Oncol. 2014, 16, 829–840. [Google Scholar] [CrossRef] [Green Version]
- Bi, W.L.; Greenwald, N.F.; Abedalthagafi, M.; Wala, J.; Gibson, W.J.; Agarwalla, P.K.; Horowitz, P.; Schumacher, S.E.; Esaulova, E.; Mei, Y.; et al. Genomic landscape of high-grade meningiomas. NPJ Genom. Med. 2017, 2, 15. [Google Scholar] [CrossRef] [Green Version]
- Huang, J.; Campian, J.L.; Gao, F.; Johanns, T.M.; Desjardins, A.; Wang-Gillam, A.; Rubin, J. A phase I/II study of nivolumab plus or minus ipilimumab in combination with multifraction stereotactic radiosurgery for recurrent high-grade radiation-relapsed meningioma. J. Clin. Oncol. 2019, 37, TPS2073. [Google Scholar] [CrossRef]
- Li, Y.D.; Veliceasa, D.; Lamano, J.B.; Lamano, J.B.; Kaur, G.; Biyashev, D.; Horbinski, C.M.; Kruser, T.J.; Bloch, O. Systemic and local immunosuppression in patients with high-grade meningiomas. Cancer Immunol. Immunother. 2019, 68, 999–1009. [Google Scholar] [CrossRef] [PubMed]
- Savardekar, A.R.; Patra, D.P.; Bir, S.; Thakur, J.D.; Mohammed, N.; Bollam, P.; Georgescu, M.-M.; Nanda, A. Differential Tumor Progression Patterns in Skull Base Versus Non–Skull Base Meningiomas: A Critical Analysis from a Long-Term Follow-Up Study and Review of Literature. World Neurosurg. 2018, 112, e74–e83. [Google Scholar] [CrossRef]
- Meling, T.R.; Da Broi, M.; Scheie, D.; Helseth, E. Meningiomas: Skull base versus non-skull base. Neurosurg. Rev. 2019, 42, 163–173. [Google Scholar] [CrossRef] [PubMed]
- Nebot-Bral, L.; Brandao, D.; Verlingue, L.; Rouleau, E.; Caron, O.; Despras, E.; El-Dakdouki, Y.; Champiat, S.; Aoufouchi, S.; Leary, A.; et al. Hypermutated tumours in the era of immunotherapy: The paradigm of personalised medicine. Eur. J. Cancer 2017, 84, 290–303. [Google Scholar] [CrossRef]
- Du, Z.; Abedalthagafi, M.; Aizer, A.A.; McHenry, A.R.; Sun, H.H.; Bray, M.-A.; Viramontes, O.; Machaidze, R.; Brastianos, P.K.; Reardon, D.A.; et al. Increased expression of the immune modulatory molecule PD-L1 (CD274) in anaplastic meningioma. Oncotarget 2014, 6, 4704–4716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Cicco, P.; Ercolano, G.; Ianaro, A. The New Era of Cancer Immunotherapy: Targeting Myeloid-Derived Suppressor Cells to Overcome Immune Evasion. Front. Immunol. 2020, 11, 1680. [Google Scholar] [CrossRef] [PubMed]
- Nayak, L.; Iwamoto, F.M.; LaCasce, A.; Mukundan, S.; Roemer, M.G.M.; Chapuy, B.; Armand, P.; Rodig, S.J.; Shipp, M.A. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017, 129, 3071–3073. [Google Scholar] [CrossRef] [Green Version]
- Schaff, L.R.; Grommes, C. Update on Novel Therapeutics for Primary CNS Lymphoma. Cancers 2021, 13, 5372. [Google Scholar] [CrossRef]
- Ostrom, Q.T.; Patil, N.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013–2017. Neuro-Oncol. 2020, 22 (Suppl. 2), iv1–iv96. [Google Scholar] [CrossRef]
- Zhang, N.; Zuo, Y.; Jiang, L.; Peng, Y.; Huang, X.; Zuo, L. Epstein-Barr Virus and Neurological Diseases. Front. Mol. Biosci. 2022, 8, 816098. [Google Scholar] [CrossRef]
- Jahnke, K.; Thiel, E.; Martus, P.; Herrlinger, U.; Weller, M.; Fischer, L.; Korfel, A.; on behalf of the German Primary Central Nervous System Lymphoma Study Group (G-PCNSL-SG). Relapse of primary central nervous system lymphoma: Clinical features, outcome and prognostic factors. J. Neuro-Oncol. 2006, 80, 159–165. [Google Scholar] [CrossRef] [PubMed]
- Tao, K.; Wang, X.; Tian, X. Relapsed Primary Central Nervous System Lymphoma: Current Advances. Front. Oncol. 2021, 11, 649789. [Google Scholar] [CrossRef] [PubMed]
- Lin, N.; Song, Y.; Zhu, J. Immune checkpoint inhibitors in malignant lymphoma: Advances and perspectives. Chin. J. Cancer Res. 2020, 32, 303–318. [Google Scholar] [CrossRef]
- Chapuy, B.; Roemer, M.G.M.; Stewart, C.; Tan, Y.; Abo, R.P.; Zhang, L.; Dunford, A.J.; Meredith, D.M.; Thorner, A.R.; Jordanova, E.S.; et al. Targetable genetic features of primary testicular and primary central nervous system lymphomas. Blood 2016, 127, 869–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grommes, C.; Nayak, L.; Tun, H.W.; Batchelor, T.T. Introduction of novel agents in the treatment of primary CNS lymphoma. Neuro-Oncol. 2018, 21, 306–313. [Google Scholar] [CrossRef]
- Ou, A.; Sumrall, A.; Phuphanich, S.; Spetzler, D.; Gatalica, Z.; Xiu, J.; Michelhaugh, S.; Brenner, A.; Pandey, M.; Kesari, S.; et al. Primary CNS lymphoma commonly expresses immune response biomarkers. Neuro-Oncol. Adv. 2020, 2, vdaa018. [Google Scholar] [CrossRef]
- Monabati, A.; Nematollahi, P.; Dehghanian, A.; Safaei, A.; Sadeghipour, A.; Movahedinia, S.; Mokhtari, M. Immune Checkpoint Molecules in Primary Diffuse Large B-Cell Lymphoma of the Central Nervous System. Basic Clin. Neurosci. J. 2020, 11, 491–498. [Google Scholar] [CrossRef]
- Berghoff, A.S.; Ricken, G.; Widhalm, G.; Rajky, O.; Hainfellner, J.A.; Birner, P.; Raderer, M.; Preusser, M. PD1 (CD279) and PD-L1 (CD274, B7H1) expression in primary central nervous system lymphomas (PCNSL). Clin. Neuropathol. 2014, 33, 42–49. [Google Scholar] [CrossRef]
- Furuse, M.; Nonoguchi, N.; Omura, N.; Shirahata, M.; Iwasaki, K.; Inui, T.; Kuroiwa, T.; Kuwabara, H.; Miyatake, S.-I. Immunotherapy of Nivolumab with Dendritic Cell Vaccination Is Effective against Intractable Recurrent Primary Central Nervous System Lymphoma: A Case Report. Neurol. Med.-Chir. 2017, 57, 191–197. [Google Scholar] [CrossRef] [Green Version]
- Panjwani, P.K.; Charu, V.; DeLisser, M.; Molina-Kirsch, H.; Natkunam, Y.; Zhao, S. Programmed death-1 ligands PD-L1 and PD-L2 show distinctive and restricted patterns of expression in lymphoma subtypes. Hum. Pathol. 2018, 71, 91–99. [Google Scholar] [CrossRef]
- Miyasato, Y.; Takashima, Y.; Takeya, H.; Yano, H.; Hayano, A.; Nakagawa, T.; Makino, K.; Takeya, M.; Yamanaka, R.; Komohara, Y. The expression of PD-1 ligands and IDO1 by macrophage/microglia in primary central nervous system lymphoma. J. Clin. Exp. Hematop. 2018, 58, 95–101. [Google Scholar] [CrossRef] [Green Version]
- Alame, M.; Pirel, M.; Costes-Martineau, V.; Bauchet, L.; Fabbro, M.; Tourneret, A.; De Oliveira, L.; Durand, L.; Roger, P.; Gonzalez, S.; et al. Characterisation of tumour microenvironment and immune checkpoints in primary central nervous system diffuse large B cell lymphomas. Virchows Arch. 2020, 476, 891–902. [Google Scholar] [CrossRef] [PubMed]
- El-Tawab, R.; Hamada, A.; Elhagracy, R.; Pinto, K.; Alshemmari, S. Promising effect of PDL1 inhibitors in the front-line management of primary aggressive central nervous system lymphoma: A case report. Hematol. Oncol. Stem Cell Ther. 2020, 20. [Google Scholar] [CrossRef]
- Graber, J.J.; Plato, B.; Mawad, R.; Moore, D.J. Pembrolizumab immunotherapy for relapsed CNS Lymphoma. Leuk. Lymphoma 2020, 61, 1766–1768. [Google Scholar] [CrossRef]
- Ambady, P.; Szidonya, L.; Firkins, J.; James, J.; Johansson, K.; White, T.; Jezierski, C.; Doolittle, N.D.; Neuwelt, E.A. Combination immunotherapy as a non-chemotherapy alternative for refractory or recurrent CNS lymphoma. Leuk. Lymphoma 2019, 60, 515–518. [Google Scholar] [CrossRef] [PubMed]
- Gavrilenko, A.N.; Volkov, N.P.; Shmidt, D.I.; Polushin, A.Y.; Kondakova, E.; Lepik, K.V.; Zalaylov, Y.R.; Popova, M.O.; Kulagin, A.D.; Afanasyev, B.V.; et al. Nivolumab in Primary CNS Lymphoma and Primary Testicular Lymphoma with CNS Involvement: Single Center Experience. Blood 2020, 136, 4. [Google Scholar] [CrossRef]
- Barnholtz-Sloan, J.S.; Sloan, A.E.; Davis, F.G.; Vigneau, F.D.; Lai, P.; Sawaya, R.E. Incidence Proportions of Brain Metastases in Patients Diagnosed (1973 to 2001) in the Metropolitan Detroit Cancer Surveillance System. J. Clin. Oncol. 2004, 22, 2865–2872. [Google Scholar] [CrossRef] [PubMed]
- Stelzer, K. Epidemiology and prognosis of brain metastases. Surg. Neurol. Int. 2013, 4, S192–S202. [Google Scholar] [CrossRef]
- Brastianos, P.K.; Carter, S.L.; Santagata, S.; Cahill, D.P.; Taylor-Weiner, A.; Jones, R.T.; Van Allen, E.M.; Lawrence, M.S.; Horowitz, P.M.; Cibulskis, K.; et al. Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets. Cancer Discov. 2015, 5, 1164–1177. [Google Scholar] [CrossRef] [Green Version]
- Orozco, J.I.J.; Knijnenburg, T.A.; Manughian-Peter, A.O.; Salomon, M.P.; Barkhoudarian, G.; Jalas, J.R.; Wilmott, J.S.; Hothi, P.; Wang, X.; Takasumi, Y.; et al. Epigenetic profiling for the molecular classification of metastatic brain tumors. Nat. Commun. 2018, 9, 4627. [Google Scholar] [CrossRef] [PubMed]
- Mansfield, A.S.; Ren, H.; Sutor, S.; Sarangi, V.; Nair, A.; Davila, J.; Elsbernd, L.R.; Udell, J.B.; Dronca, R.S.; Park, S.; et al. Contraction of T cell richness in lung cancer brain metastases. Sci. Rep. 2018, 8, 2171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fischer, G.M.; Jalali, A.; Kircher, D.A.; Lee, W.-C.; McQuade, J.L.; Haydu, L.E.; Joon, A.Y.; Reuben, A.; de Macedo, M.P.; Carapeto, F.C.L.; et al. Molecular Profiling Reveals Unique Immune and Metabolic Features of Melanoma Brain Metastases. Cancer Discov. 2019, 9, 628–645. [Google Scholar] [CrossRef] [Green Version]
- Fukumura, K.; Malgulwar, P.B.; Fischer, G.M.; Hu, X.; Mao, X.; Song, X.; Hernandez, S.D.; Zhang, X.H.-F.; Zhang, J.; Parra, E.R.; et al. Multi-omic molecular profiling reveals potentially targetable abnormalities shared across multiple histologies of brain metastasis. Acta Neuropathol. 2021, 141, 303–321. [Google Scholar] [CrossRef]
- Alvarez-Breckenridge, C.; Remon, J.; Piña, Y.; Nieblas-Bedolla, E.; Forsyth, P.; Hendriks, L.; Brastianos, P.K. Emerging Systemic Treatment Perspectives on Brain Metastases: Moving Toward a Better Outlook for Patients. Am. Soc. Clin. Oncol. Educ. Book 2022, 42, 147–165. [Google Scholar] [CrossRef]
- Tawbi, H.A.; Forsyth, P.A.; Algazi, A.; Hamid, O.; Hodi, F.S.; Moschos, S.J.; Khushalani, N.I.; Lewis, K.; Lao, C.D.; Postow, M.A.; et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. N. Engl. J. Med. 2018, 379, 722–730. [Google Scholar] [CrossRef] [PubMed]
- Margolin, K.; Ernstoff, M.S.; Hamid, O.; Lawrence, D.; McDermott, D.; Puzanov, I.; Wolchok, J.D.; Clark, J.I.; Sznol, M.; Logan, T.F.; et al. Ipilimumab in patients with melanoma and brain metastases: An open-label, phase 2 trial. Lancet Oncol. 2012, 13, 459–465. [Google Scholar] [CrossRef] [PubMed]
- A Tawbi, H.; A Forsyth, P.; Hodi, F.S.; Lao, C.D.; Moschos, S.J.; Hamid, O.; Atkins, M.B.; Lewis, K.; Thomas, R.P.; A Glaspy, J.; et al. Safety and efficacy of the combination of nivolumab plus ipilimumab in patients with melanoma and asymptomatic or symptomatic brain metastases (CheckMate 204). Neuro-Oncol. 2021, 23, 1961–1973. [Google Scholar] [CrossRef]
- Long, G.V.; Atkinson, V.; Lo, S.; Sandhu, S.; Guminski, A.D.; Brown, M.P.; Wilmott, J.S.; Edwards, J.; Gonzalez, M.; Scolyer, R.A.; et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: A multicentre randomised phase 2 study. Lancet Oncol. 2018, 19, 672–681. [Google Scholar] [CrossRef] [PubMed]
- A Tawbi, H.; A Forsyth, P.; Hodi, F.S.; Algazi, A.P.; Hamid, O.; Lao, C.D.; Moschos, S.J.; Atkins, M.B.; Lewis, K.; A Postow, M.; et al. Long-term outcomes of patients with active melanoma brain metastases treated with combination nivolumab plus ipilimumab (CheckMate 204): Final results of an open-label, multicentre, phase 2 study. Lancet Oncol. 2021, 22, 1692–1704. [Google Scholar] [CrossRef]
- Di Giacomo, A.M.; A Ascierto, P.; Pilla, L.; Santinami, M.; Ferrucci, P.F.; Giannarelli, D.; Marasco, A.; Rivoltini, L.; Simeone, E.; Nicoletti, S.V.; et al. Ipilimumab and fotemustine in patients with advanced melanoma (NIBIT-M1): An open-label, single-arm phase 2 trial. Lancet Oncol. 2012, 13, 879–886. [Google Scholar] [CrossRef]
- Di Giacomo, A.M.; Chiarion-Sileni, V.; Del Vecchio, M.; Ferrucci, P.F.; Guida, M.; Quaglino, P.; Guidoboni, M.; Marchetti, P.; Cutaia, O.; Amato, G.; et al. Primary Analysis and 4-Year Follow-Up of the Phase III NIBIT-M2 Trial in Melanoma Patients With Brain Metastases. Clin. Cancer Res. 2021, 27, 4737–4745. [Google Scholar] [CrossRef]
- Goldberg, S.B.; Gettinger, S.N.; Mahajan, A.; Chiang, A.C.; Herbst, R.S.; Sznol, M.; Tsiouris, A.J.; Cohen, J.; Vortmeyer, A.; Jilaveanu, L.; et al. Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: Early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 976–983. [Google Scholar] [CrossRef] [Green Version]
- Kluger, H.M.; Chiang, V.; Mahajan, A.; Zito, C.R.; Sznol, M.; Tran, T.; Weiss, S.A.; Cohen, J.V.; Yu, J.; Hegde, U.; et al. Long-Term Survival of Patients With Melanoma With Active Brain Metastases Treated With Pembrolizumab on a Phase II Trial. J. Clin. Oncol. 2018, 37, 52–60. [Google Scholar] [CrossRef] [PubMed]
- Ascierto, P.A.; Del Vecchio, M.; Robert, C.; Mackiewicz, A.; Chiarion-Sileni, V.; Arance, A.; Lebbé, C.; Bastholt, L.; Hamid, O.; Rutkowski, P.; et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: A randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017, 18, 611–622. [Google Scholar] [CrossRef]
- Ascierto, P.A.; Del Vecchio, M.; Mackiewicz, A.; Robert, C.; Chiarion-Sileni, V.; Arance, A.; Lebbé, C.; Svane, I.M.; McNeil, C.; Rutkowski, P.; et al. Overall survival at 5 years of follow-up in a phase III trial comparing ipilimumab 10 mg/kg with 3 mg/kg in patients with advanced melanoma. J. Immunother. Cancer 2020, 8, e000391. [Google Scholar] [CrossRef] [PubMed]
- Pelster, M.S.; Amaria, R.N. Combined targeted therapy and immunotherapy in melanoma: A review of the impact on the tumor microenvironment and outcomes of early clinical trials. Ther. Adv. Med Oncol. 2019, 11, 1758835919830826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsao, M.; Xu, W.; Sahgal, A. A meta-analysis evaluating stereotactic radiosurgery, whole-brain radiotherapy, or both for patients presenting with a limited number of brain metastases. Cancer 2012, 118, 2486–2493. [Google Scholar] [CrossRef]
- Tallet, A.V.; Azria, D.; Barlesi, F.; Spano, J.-P.; Carpentier, A.F.; Gonçalves, A.; Metellus, P. Neurocognitive function impairment after whole brain radiotherapy for brain metastases: Actual assessment. Radiat. Oncol. 2012, 7, 77. [Google Scholar] [CrossRef] [Green Version]
- Krummel, D.A.P.; Nasti, T.H.; Izar, B.; Press, R.H.; Xu, M.; Lowder, L.; Kallay, L.; Rupji, M.; Rosen, H.; Su, J.; et al. Impact of Sequencing Radiation Therapy and Immune Checkpoint Inhibitors in the Treatment of Melanoma Brain Metastases. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 157–163. [Google Scholar] [CrossRef] [Green Version]
- Weil, R.J.; Palmieri, D.C.; Bronder, J.L.; Stark, A.M.; Steeg, P.S. Breast Cancer Metastasis to the Central Nervous System. Am. J. Pathol. 2005, 167, 913–920. [Google Scholar] [CrossRef] [Green Version]
- Sperduto, P.W.; Kased, N.; Roberge, D.; Chao, S.T.; Shanley, R.; Luo, X.; Sneed, P.K.; Suh, J.; Weil, R.J.; Jensen, A.W.; et al. The effect of tumor subtype on the time from primary diagnosis to development of brain metastases and survival in patients with breast cancer. J. Neuro-Oncol. 2013, 112, 467–472. [Google Scholar] [CrossRef]
- Schmid, P.; Adams, S.; Rugo, H.S.; Schneeweiss, A.; Barrios, C.H.; Iwata, H.; Diéras, V.; Hegg, R.; Im, S.-A.; Shaw Wright, G.; et al. Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N. Engl. J. Med. 2018, 379, 2108–2121. [Google Scholar] [CrossRef] [PubMed]
- Cortes, J.; Cescon, D.W.; Rugo, H.S.; Nowecki, Z.; Im, S.-A.; Yusof, M.M.; Gallardo, C.; Lipatov, O.; Barrios, C.H.; Holgado, E.; et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet 2020, 396, 1817–1828. [Google Scholar] [CrossRef]
- Berghoff, A.S.; Fuchs, E.; Ricken, G.; Mlecnik, B.; Bindea, G.; Spanberger, T.; Hackl, M.; Widhalm, G.; Dieckmann, K.; Prayer, D.; et al. Density of tumor-infiltrating lymphocytes correlates with extent of brain edema and overall survival time in patients with brain metastases. Oncoimmunology 2016, 5, e1057388. [Google Scholar] [CrossRef]
- Waqar, S.N.; Samson, P.P.; Robinson, C.G.; Bradley, J.; Devarakonda, S.; Du, L.; Govindan, R.; Gao, F.; Puri, V.; Morgensztern, D. Non–small-cell Lung Cancer With Brain Metastasis at Presentation. Clin. Lung Cancer 2018, 19, e373–e379. [Google Scholar] [CrossRef]
- Shi, A.A.; Digumarthy, S.R.; Temel, J.S.; Halpern, E.F.; Kuester, L.B.; Aquino, S.L. Does Initial Staging or Tumor Histology Better Identify Asymptomatic Brain Metastases in Patients with Non–small Cell Lung Cancer? J. Thorac. Oncol. 2006, 1, 205–210. [Google Scholar] [CrossRef] [Green Version]
- Sorensen, J.B.; Hansen, H.H.; Hansen, M.; Dombernowsky, P. Brain Metastases in Adenocarcinoma of the Lung: Frequency, Risk Groups, and Prognosis. J. Clin. Oncol. 1988, 6, 1474–1480. [Google Scholar] [CrossRef] [PubMed]
- Kelly, K.; A Bunn, P. Is it time to reevaluate our approach to the treatment of brain metastases in patients with non-small cell lung cancer? Lung Cancer 1998, 20, 85–91. [Google Scholar] [CrossRef]
- Patil, T.; Smith, D.E.; Bunn, P.A.; Aisner, D.L.; Le, A.T.; Hancock, M.; Purcell, W.T.; Bowles, D.W.; Camidge, D.R.; Doebele, R.C. The Incidence of Brain Metastases in Stage IV ROS1-Rearranged Non–Small Cell Lung Cancer and Rate of Central Nervous System Progression on Crizotinib. J. Thorac. Oncol. 2018, 13, 1717–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rangachari, D.; Yamaguchi, N.; VanderLaan, P.A.; Folch, E.; Mahadevan, A.; Floyd, S.R.; Uhlmann, E.J.; Wong, E.T.; Dahlberg, S.E.; Huberman, M.S.; et al. Brain metastases in patients with EGFR -mutated or ALK -rearranged non-small-cell lung cancers. Lung Cancer 2015, 88, 108–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peters, S.; Camidge, D.R.; Shaw, A.T.; Gadgeel, S.; Ahn, J.S.; Kim, D.W.; Ou, S.H.I.; Pérol, M.; Dziadziuszko, R.; Rosell, R.; et al. Alectinib versus Crizotinib in Untreated ALK-Positive Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 377, 829–838. [Google Scholar] [CrossRef]
- Reungwetwattana, T.; Nakagawa, K.; Cho, B.C.; Cobo, M.; Cho, E.K.; Bertolini, A.; Bohnet, S.; Zhou, C.; Lee, K.H.; Nogami, N.; et al. CNS Response to Osimertinib Versus Standard Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients With Untreated EGFR-Mutated Advanced Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3290–3297. [Google Scholar] [CrossRef]
- Urbanska, E.M.; Santoni-Rugiu, E.; Melchior, L.C.; Carlsen, J.F.; Sørensen, J.B. Intracranial Response of ALK+ Non-Small-cell Lung Cancer to Second-line Dose-escalated Brigatinib After Alectinib Discontinuation Due to Drug-induced Hepatitis and Relapse After Whole Brain Radiotherapy Followed by Stereotactic Radiosurgery. Clin. Lung Cancer 2020, 22, e528–e532. [Google Scholar] [CrossRef]
- Lin, J.J.; Jiang, G.Y.; Joshipura, N.; Ackil, J.; Digumarthy, S.R.; Rincon, S.P.; Yeap, B.Y.; Gainor, J.F.; Shaw, A.T. Efficacy of Alectinib in Patients with ALK-Positive NSCLC and Symptomatic or Large CNS Metastases. J. Thorac. Oncol. 2019, 14, 683–690. [Google Scholar] [CrossRef]
- Facchinetti, F.; Levy, A.; Ammari, S.; Naltet, C.; Lavaud, P.; Aldea, M.; Vasseur, D.; Planchard, D.; Besse, B. Meningeal “Lazarus Response” to Lorlatinib in a ROS1-Positive NSCLC Patient Progressing to Entrectinib. Cancer Manag. Res. 2021, 13, 2805–2810. [Google Scholar] [CrossRef]
- Hochmair, M.; Weinlinger, C.; Prosch, H. Intracranial remission with brigatinib rechallenge as fifth-line ALK inhibition therapy in a lung cancer patient. Anti-Cancer Drugs 2019, 30, 1058–1060. [Google Scholar] [CrossRef] [PubMed]
- Koba, T.; Kijima, T.; Takimoto, T.; Hirata, H.; Naito, Y.; Hamaguchi, M.; Otsuka, T.; Kuroyama, M.; Nagatomo, I.; Takeda, Y.; et al. Rapid intracranial response to osimertinib, without radiotherapy, in nonsmall cell lung cancer patients harboring the EGFR T790M mutation two case reports. Medicine 2017, 96, e6087. [Google Scholar] [CrossRef] [PubMed]
- Molinier, O.; Besse, B.; Barlesi, F.; Audigier-Valette, C.; Friard, S.; Monnet, I.; Jeannin, G.; Mazières, J.; Cadranel, J.; Hureaux, J.; et al. IFCT-1502 CLINIVO: Real-world evidence of long-term survival with nivolumab in a nationwide cohort of patients with advanced non-small-cell lung cancer. ESMO Open 2022, 7, 100353. [Google Scholar] [CrossRef] [PubMed]
- Mansfield, A.S.; Herbst, R.S.; de Castro, G.; Hui, R.; Peled, N.; Kim, D.-W.; Novello, S.; Satouchi, M.; Wu, Y.-L.; Garon, E.B.; et al. Outcomes With Pembrolizumab Monotherapy in Patients With Programmed Death-Ligand 1–Positive NSCLC With Brain Metastases: Pooled Analysis of KEYNOTE-001, 010, 024, and 042. JTO Clin. Res. Rep. 2021, 2, 100205. [Google Scholar] [CrossRef]
- Chen, H.; Feng, Y.; Zhou, Y.; Tao, Y.; Tang, L.; Shi, Y. Brain metastases and immune checkpoint inhibitors in non-small cell lung cancer: A systematic review and meta-analysis. Cancer Immunol. Immunother. 2022, 71, 3071–3085. [Google Scholar] [CrossRef]
- Camidge, D.R.; Lee, E.Q.; Lin, N.U.; Margolin, K.; Ahluwalia, M.S.; Bendszus, M.; Chang, S.M.; Dancey, J.; E de Vries, E.G.; Harris, G.J.; et al. Clinical trial design for systemic agents in patients with brain metastases from solid tumours: A guideline by the Response Assessment in Neuro-Oncology Brain Metastases working group. Lancet Oncol. 2018, 19, e20–e32. [Google Scholar] [CrossRef]
- Schoenmaekers, J.J.A.O.; Dursun, S.; Biesmans, C.; De Ruysscher, D.K.M.; Broen, M.P.G.; Remon, J.; Dingemans, A.-M.C.; Hendriks, L.E.L. Dynamics of eligibility criteria for central nervous system metastases in non-small cell lung cancer randomized clinical trials over time: A systematic review. Crit. Rev. Oncol./Hematol. 2021, 166, 103460. [Google Scholar] [CrossRef]
- El Rassy, E.; Botticella, A.; Kattan, J.; Le Péchoux, C.; Besse, B.; Hendriks, L. Non-small cell lung cancer brain metastases and the immune system: From brain metastases development to treatment. Cancer Treat. Rev. 2018, 68, 69–79. [Google Scholar] [CrossRef] [Green Version]
- Vilariño, N.; Bruna, J.; Bosch-Barrera, J.; Valiente, M.; Nadal, E. Immunotherapy in NSCLC patients with brain metastases. Understanding brain tumor microenvironment and dissecting outcomes from immune checkpoint blockade in the clinic. Cancer Treat. Rev. 2020, 89, 102067. [Google Scholar] [CrossRef] [PubMed]
- Crinò, L.; Bronte, G.; Bidoli, P.; Cravero, P.; Minenza, E.; Cortesi, E.; Garassino, M.C.; Proto, C.; Cappuzzo, F.; Grossi, F.; et al. Nivolumab and brain metastases in patients with advanced non-squamous non-small cell lung cancer. Lung Cancer 2019, 129, 35–40. [Google Scholar] [CrossRef] [Green Version]
- Goldberg, S.B.; Schalper, K.A.; Gettinger, S.N.; Mahajan, A.; Herbst, R.S.; Chiang, A.C.; Lilenbaum, R.; Wilson, F.H.; Omay, S.B.; Yu, J.B.; et al. Pembrolizumab for management of patients with NSCLC and brain metastases: Long-term results and biomarker analysis from a non-randomised, open-label, phase 2 trial. Lancet Oncol. 2020, 21, 655–663. [Google Scholar] [CrossRef]
- Hellmann, M.D.; Paz-Ares, L.; Bernabe Caro, R.; Zurawski, B.; Kim, S.-W.; Carcereny Costa, E.; Park, K.; Alexandru, A.; Lupinacci, L.; de la Mora Jimenez, E.; et al. Nivolumab plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2019, 381, 2020–2031. [Google Scholar] [CrossRef] [PubMed]
- Gandhi, L.; Rodríguez-Abreu, D.; Gadgeel, S.; Esteban, E.; Felip, E.; De Angelis, F.; Domine, M.; Clingan, P.; Hochmair, M.J.; Powell, S.F.; et al. Pembrolizumab plus Chemotherapy in Metastatic Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2018, 378, 2078–2092. [Google Scholar] [CrossRef] [PubMed]
- Gadgeel, S.; Rodríguez-Abreu, D.; Speranza, G.; Esteban, E.; Felip, E.; Dómine, M.; Hui, R.; Hochmair, M.J.; Clingan, P.; Powell, S.F.; et al. Updated Analysis From KEYNOTE-189: Pembrolizumab or Placebo Plus Pemetrexed and Platinum for Previously Untreated Metastatic Nonsquamous Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2020, 38, 1505–1517. [Google Scholar] [CrossRef]
- Rittmeyer, A.; Barlesi, F.; Waterkamp, D.; Park, K.; Ciardiello, F.; von Pawel, J.; Gadgeel, S.M.; Hida, T.; Kowalski, D.M.; Dols, M.C.; et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): A phase 3, open-label, multicentre randomised controlled trial. Lancet 2017, 389, 255–265. [Google Scholar] [CrossRef]
- Brastianos, P.K.; Strickland, M.R.; Lee, E.Q.; Wang, N.; Cohen, J.V.; Chukwueke, U.; Forst, D.A.; Eichler, A.; Overmoyer, B.; Lin, N.U.; et al. Phase II study of ipilimumab and nivolumab in leptomeningeal carcinomatosis. Nat. Commun. 2021, 12, 5954. [Google Scholar] [CrossRef]
- Brastianos, P.K.; Lee, E.Q.; Cohen, J.V.; Tolaney, S.M.; Lin, N.U.; Wang, N.; Chukwueke, U.; White, M.D.; Nayyar, N.; Kim, A.; et al. Single-arm, open-label phase 2 trial of pembrolizumab in patients with leptomeningeal carcinomatosis. Nat. Med. 2020, 26, 1280–1284. [Google Scholar] [CrossRef] [PubMed]
- Grossman, S.A.; Krabak, M.J. Leptomeningeal Carcinomatsis. Cancer Treat Rev. 1999, 25, 103–119. [Google Scholar] [CrossRef] [PubMed]
- Le Rhun, E.; Preusser, M.; Bent, M.V.D.; Andratschke, N.; Weller, M. How we treat patients with leptomeningeal metastases. ESMO Open 2019, 4, e000507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beauchesne, P. Intrathecal chemotherapy for treatment of leptomeningeal dissemination of metastatic tumours. Lancet Oncol. 2010, 11, 871–879. [Google Scholar] [CrossRef]
- Iorgulescu, J.B.; Gokhale, P.C.; Speranza, M.C.; Eschle, B.K.; Poitras, M.J.; Wilkens, M.K.; Soroko, K.M.; Chhoeu, C.; Knott, A.; Gao, Y.; et al. Concurrent Dexamethasone Limits the Clinical Benefit of Immune Checkpoint Blockade in Glioblastoma. Clin. Cancer Res. 2021, 27, 276–287. [Google Scholar] [CrossRef]
Author | Phase | Patients | Trial Arms | Outcomes |
---|---|---|---|---|
KeyNote-028 [13] | Ib | 26 patients with PD-L1-positive recurrent GBM | Pembrolizumab | Investigation-assessed ORR (by RECIST v1.1): 8%. DOR: 8.3 and 22.8 months in 2 patients, respectively; mPFS: 2.8 months; PFS6 rate: 37.7% mOS: 13.1 months; OS rate at 12 months: 58% |
Nayak L et al., 2021 [17] | II | 80 patients with recurrent GBM | A: Pembrolizumab + Bevacizumab B: Pembrolizumab | A: 20% ORR, 26% PFS-6 rate;mOS: 8.8 months; B: 0% ORR |
Nayak et al., 2022 [18] | II | 137 patients with newly diagnosed and recurrent GBM | A: Durvalumab + RT B: Durvalumab B2: Durvalumab + Bevacizumab B3 and C: Durvalumab + low-dose Bevacizumab | Primary endpoints: A: OS-12 (not met) B, B2, B3: PFS-6 (not met) C: OS-6 (not met) |
Lukas R et al., 2018 [19] | Ia | 16 patients with recurrent GBM | Atezolizumab | Treatment safe and tolerated. 6% ORR; mPFS: 1.2 months; mOS: 4.2 months; |
Chiocca EA et al. 2021 [20] | I | 21 patients with recurrent GBM | A: Nivolumab 1 mg/kg + 10 mg VDX; B: Nivolumab 3 mg/kg + 10 mg VDX C: Nivolumab 3 mg/kg + 20 mg VDX | Treatment safe and tolerated. A and B: mOS: 16.9 months; C: mOS: 8.5 months |
Omuro et al., 2022 (CheckMate-143) [21] | I | 136 patients with newly diagnosed GBM | A: Nivolumab + TMZ + RT B: Nivolumab + RT | Primary endpoints: safety and tolerability (met; more frequent lymphopenia in cohort A); 2ry endpoint: OS (similar between A and B, different according to MGMT methylation status); Exploratory endpoints: PFS |
CheckMate 498 [22] | III | 560 patients with newly diagnosed MGMTunm GBM | A. Nivolumab + RT B: TMZ + RT | Treatment safe and tolerated. A: mOS: 13.4 months; PFS: 6 months; grade 3/4 AEs: 21.9%; serious AEs: 17.3%; B: mOS: 14.9 months; PFS: 6.2 months; grade 3/4 AEs; 25.1%; serious AEs: 7.6% |
Schalper K et al., 2019 [23] | II | 30 patients with resectable GBM (twenty-seven recurrent and three newly diagnosed) | Neoadjuvant Nivolumab | Safe and tolerable treatment. mPFS: 4.1 months; mOS: 7.3 months |
Cloughesy T et al., 2019 [24] | NA | 32 patients with recurrent GBM (53% MGMTm, 34% MGMTunm, and 13% unknown) | A: Neoadjuvant + adjuvant Pembrolizumab B. adjuvant Pembrolizumab | Treatment safe and tolerated. mOS: 13.7 vs. 7.5 months (A vs. B); mPFS: 3.3 vs. 2.4 months (A vs. B) |
Reardon et al., 2020 [25] | III | 369 with recurrent GBM (23.4% MGMTm, 22.7% MGMTunm, and 36.2% unknown) | A: Nivolumab B: Bevacizumab | Primary endpoint not met. Nivolumab safe and tolerated. A: mOS: 9.8 months; ORR: 7.8%; B: mOS: 10 months; ORR: 23.1%; 1-yr OS: 42% for both groups;grade 3/4 AEs similar in A and B |
CheckMate 548 [26] | III | 716 patients with newly diagnosed MGMTm GBM | A: Nivolumab + TMZ + RT B: PBO + TMZ +RT | Treatment safe and tolerated. mPFS: 10.6 vs. 10.3 months (A vs. B); mOS: 28.9 vs. 32.1 months (A vs. B); with basal corticosteroids, mOS: 31.3 vs. 33 months (A vs. B) |
Checkate-143 [22] | I | 40 patients with recurrent GBM | A: Nivolumab 3mg/kg B. Ipilimumab 3 mg/kg + Nivolumab 1 mg/kg C: Ipilimumab 1mg/kg+ Nivolumab 3 mg/kg | Nivolumab monotherapy better tolerated than Ipilimumab-nivolumab combo; mPFS: 1.9 vs. 1.5 vs. 2.1 months (A vs. B vs. C); mOS: 10.4 vs. 9.2 vs. 7.3 months (A vs. B vs. C) |
Author or Trial Name | Phase | Drug | Cohort | Results |
---|---|---|---|---|
Furuse et al., 2017 [69] | Case report | Nivolumab + DC vaccination | 1 patient | CR maintained for 10 months |
Nayak et al., 2017 [57] | Case series | Nivolumab | 5 patietns (4 with PCNSL and 1 with PTL) | 4 patients with CR and 1 with PR; |
Graber J et al., 2020 [74] | Case series | Pembrolizumab | 5 patients (PCNSL and SCNSL) | Prolonged remission in 3 out of 5 patients |
Ambady et al., 2019 [75] | Retrospective study | Nivolumab/Pembrolizumab and Rituximab | 6 patients (three with PCNSL and three SCNSL) | 3 out of 6 patients with CR |
Gavrilenko A et al., 2020 [75,76]; | Case series | Nivolumab | 8 patients with PCNSL and one with PTL | 2-year OS: 44%; mOS: 12 months; 2-year PFS: 26%; mPFS: 12 months; |
Study/Studies | Phase | Therapies | Patient Cohort(s) | Results (ORR, PFS, OS) |
---|---|---|---|---|
Margolin et al., 2012 [86] | II | Ipilimumab | Cohort A: asymptomatic MBM (51); Cohort B: symptomatic MBM on (21) | iDCR: 24 vs. 10% (A vs. B); mPFS: 1.5–1.9 vs. 1.2 months (A vs. B); mOS: 7 vs. 3.7 months (A vs. B) |
NIBIT M1 [90]; | II | Ipilimumab + Fotemustine | 20 patients with asymptomatic brain metastases out of a cohort of eight-six patients with advanced melanoma | In MBM patients: Brain-PFS: 3 months |
NIBIT-M2 [91] | III | Ipilimumab, Nivolumab, Fotemustine | 80 patients with MBM Arm A: Fotemustine (27) Arm B: Ipilimumab + Fotemustine (26) Arm C: Ipilimumab + Nivolumab (27) | mOS: 8.5 vs. 8.2 vs. 29.2 months (A vs. B vs. C); mPFS: 3 vs. 3 vs. 8.7 months (A vs. B vs. C); ICR: 0 vs. 19.2 vs. 44.4% (A vs. B vs. C) |
Goldberg et al., 2016 [92] | II | Pembrolizumab | 18 patients with MBM in a cohort of 52 patients with brain metastases | ORR: 22% mOS: not reached |
Kluger et al., 2018; [93] | II | Pembrolizumab | 23 patients with MBM | RR: 26% mPFS: 2 months; mOS: 17 months |
Ascierto et al., 2017,2020 [94,95] | III | Ipilimumab A: 10 mg/kg; B: 3 mg/kg; | 127 patients with MBMs in a cohort of 727 patients with advanced melanoma | In MBM patients: mOS: 7 months vs. 5.7 months (A vs. B) |
Checkmate204 [89] | II | Nivolumab + Ipilimumab | 119 patients with MBM Cohort A: asymptomatic (101); Cohort B: symptomatic (18) | iORR: 53.5% vs. 16.7% (A vs. B); 36-month iPFS: 54.1% vs. 18.9% (A vs. B); 36-month OS: 71.9% vs. 36.6% (A vs. B) |
ABC study [88] | II | Nivolumab + Ipilimumab vs. Nivolumab | 79 patients with MBM; Cohort A: Nivolumab + Ipilimumab (36) Cohort B: Nivolumab (27); Cohort C: Nivolumab prior Tx, symptomatic, or with LM (16); | ICR 46% vs. 20% vs. 6% (A vs. B vs. C); ICCR 17% vs. 12% vs. 0% (A vs. B vs. C); iPFS: not reached vs. 2.5 vs. 2.3 months (A vs. B vs. C); OS: not reached vs. 18.5 vs. 5.1 months (A vs. B vs. C) |
Study | Phase | Therapy | Patient Cohort | Patients with BM | Results |
---|---|---|---|---|---|
Goldberg et al., 2020 [126] | II | Pembrolizumab | 42 asymptomatic patients with untreated BM from NSCLC; Cohort A: PD-L1 expression ≥1%; Cohort B: PD-L1 expression <1% or unevaluable; | 100% | Cohort A: 29.7% BM response rate; Cohort 2: no response |
Keynote-189 study [128,129] | III | ICI Arm (A): Pembrolizumab + Pemetrexed + a Pt-based CT; Control Arm (B): Pemetrexed + a Pt-based CT; | 108 patents among a cohort of six hundred and sixteen patients with metastatic n-sq-NSCLC | 17.53% | For BM patients: mOS: 19.2 vs. 7.5 months (A vs. B); HR for OS (A vs. B): 0.41 |
Crinò et al., 2019 [125] | EAP | Nivolumab | 409 patients with asymptomatic or controlled BM in a cohort of 1588 patients with advanced n-sq-NSCLC | 26% | BM patients: mOS: 8.6 months; mPFS: 3 months; iDCR: 40%; ORR:17% |
OAK trial [130] | III | ICI Arm (A): Atezolizumab; Control Arm (B): docetaxel | 85 patients with BM in a cohort of 850 patients with previously treated stage IIIB/IV NSCLC; | 10% | For BM patients: mOS: 20.1 vs. 11.9 months (A vs. B); OS HR for Atezolizumab: 0.54 |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Alimonti, P.; Gonzalez Castro, L.N. The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors. Antibodies 2023, 12, 27. https://doi.org/10.3390/antib12020027
Alimonti P, Gonzalez Castro LN. The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors. Antibodies. 2023; 12(2):27. https://doi.org/10.3390/antib12020027
Chicago/Turabian StyleAlimonti, Paolo, and L. Nicolas Gonzalez Castro. 2023. "The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors" Antibodies 12, no. 2: 27. https://doi.org/10.3390/antib12020027
APA StyleAlimonti, P., & Gonzalez Castro, L. N. (2023). The Current Landscape of Immune Checkpoint Inhibitor Immunotherapy for Primary and Metastatic Brain Tumors. Antibodies, 12(2), 27. https://doi.org/10.3390/antib12020027