Systematic Reversal of Drug Resistance in Cancer
Abstract
:1. Introduction
2. Ecology and Evolution of Drug Resistance
2.1. The Source of Drug Resistance: Tumor Heterogeneity
2.2. The Breeding Grounds of Drug Resistance: The Intratumoral Physiological Barrier
2.2.1. Blood–Lymph Network
2.2.2. Extracellular Matrix
2.3. The Safe Havens of Drug Resistance: The Blood–Brain Barrier/Blood–Brain Tumor Barrier
3. Overcoming Drug Resistance
3.1. Managing the Source of Drug Resistance
3.1.1. Management of Cancer Evolution and Early Intervention
Cancer Type | Stage | Sample | Technique | Genetic Mutations | Ref. |
---|---|---|---|---|---|
Colorectal cancer | Advanced | Plasma | BEAMing | APC, KRAS, PIK3CA, TP53 | [89] |
Early to advanced | Plasma | ME-PCR | KRAS | [90] | |
Colorectal and breast cancer | Advanced | Plasma | WGS | Chromosomal alterations | [91] |
Ovarian cancer | Advanced | Plasma | TAm-Seq Digital PCR | TP53, PTEN, EGFR, BRAF, KRAS | [92] |
Early to advanced | Serum | Fluorescent-PCR | PIK3CA | [93] | |
Hepatocellular carcinoma | Early | Plasma | WGS | SNV | [94] |
Non-small-cell lung cancer | Advanced | Plasma | ARMS-qPCR | KRAS | [95] |
Breast and osteosarcoma | Advanced | Plasma and serum | Nested-real time PCR | Genomic alterations | [96] |
Breast cancer | Advanced | Plasma | TAm-Seq and digital PCR | PIK3CA, TP53, structural variation | [97] |
Early to advanced | Plasma | BEAMing | PIK3CA | [98] |
Cancer Type | Genetic Mutations | Subclonal Mutation | Acquired Drug Resistance | Corresponding Strategies | Ref. |
---|---|---|---|---|---|
Colorectal cancer | KRAS | Codon 12, 13, and 61 | Cetuximab | Combination therapies targeting at least two different pathways for subclonal mutations. | [82] |
Q61H, G13D, G12D | Cetuximab | Blood-based non-invasive monitoring of KRAS mutant clones and early combined treatment with an MEK inhibitor. | [83] | ||
NRAS | Positions 12 and 61 | Cetuximab Panitumumab | An EGFR-MEK concomitant blockade (cetuximab and pimasertib) induced prolonged ERK inhibition. | [74,83] | |
BRAF | Position 600 | ||||
NSCLC | EGFR | T790M | Gefitinib Erlotinib | Use irreversible inhibitors that bind covalently or reversible inhibitors that bind with a sufficient affinity to outcompete ATP. | [87,99] |
T790M mutation and MET amplification | Gefinitib Erlotinib | Combined PI3K and MEK inhibitors, T790M-specific EGFR inhibitors, or combinations of anti-EGFR therapies. | [84] | ||
PIK3CA | E545K | Paclitaxel | Paclitaxel combined with mTOR inhibitors (rapamycin or its analogues) | [99,100] | |
ALK | C1156Y, L1196M | Crizotinib | Determine the crystal structure of the ALK kinase domain with C1156Y or L1196M mutations and develop new-generation ALK inhibitors | [86] | |
KRAS | G12C | Adagrasib Sotorasib | Combined inhibition of KRASG12C and mTORC1 (RMC-4998 and RMC-6272, RM-018 and RMC-6272) | [101] | |
Melanoma | MEK1 | C121S | Vemurafenib | Gene amplification or altered pharmacokinetics: dose escalation; develop new drugs to circumvent on-target kinase resistance mechanisms. | [80] |
PDGFRβ upregulation and NRAS mutations | Q61K | Vemurafenib | Stratify patients with drug-resistant relapses and combine MEK activation inhibitors and kinase activity inhibitors. | [85] | |
V600E BRAF | Amplification | Vemurafenib | Combination with MEK1/2 inhibitor (Selumetinib) | [102] |
3.1.2. Combined with Subclonal Mutation Inhibitors
3.1.3. Model Prediction
3.1.4. Cancer Vaccines and Adoptive T-Cell Therapy
3.2. Dismantle the Breeding Grounds of Drug Resistance
3.2.1. Normalization of Blood–Lymph Network
3.2.2. Normalization of Extracellular Matrix
3.3. Breaking the Safe Havens of Drug Resistance
3.3.1. Biochemical Therapies
3.3.2. Physical Therapies
4. Conclusions and Prospectives
Author Contributions
Funding
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Sansregret, L.; Vanhaesebroeck, B.; Swanton, C. Determinants and clinical implications of chromosomal instability in cancer. Nat. Rev. Clin. Oncol. 2018, 15, 139–150. [Google Scholar] [CrossRef] [PubMed]
- Stephens, P.J.; Greenman, C.D.; Fu, B.; Yang, F.; Bignell, G.R.; Mudie, L.J.; Pleasance, E.D.; Lau, K.W.; Beare, D.; Stebbings, L.A.; et al. Massive Genomic Rearrangement Acquired in a Single Catastrophic Event during Cancer Development. Cell 2011, 144, 27–40. [Google Scholar] [CrossRef] [PubMed]
- Greaves, M. Evolutionary determinants of cancer. Cancer Discov. 2015, 5, 806–820. [Google Scholar] [CrossRef]
- Johnson, B.E.; Mazor, T.; Hong, C.; Barnes, M.; Aihara, K.; McLean, C.Y.; Fouse, S.D.; Yamamoto, S.; Ueda, H.; Tatsuno, K.; et al. Mutational Analysis Reveals the Origin and Therapy-Driven Evolution of Recurrent Glioma. Science 2014, 343, 189–193. [Google Scholar] [CrossRef]
- Vo, J.N.; Wu, Y.-M.; Mishler, J.; Hall, S.; Mannan, R.; Wang, L.; Ning, Y.; Zhou, J.; Hopkins, A.C.; Estill, J.C.; et al. The genetic heterogeneity and drug resistance mechanisms of relapsed refractory multiple myeloma. Nat. Commun. 2022, 13, 3750. [Google Scholar] [CrossRef]
- Fischer, E.S.; Böhm, K.; Lydeard, J.R.; Yang, H.; Stadler, M.B.; Cavadini, S.; Nagel, J.; Serluca, F.; Acker, V.; Lingaraju, G.M.; et al. Structure of the DDB1–CRBN E3 ubiquitin ligase in complex with thalidomide. Nature 2014, 512, 49–53. [Google Scholar] [CrossRef]
- Krönke, J.; Udeshi, N.D.; Narla, A.; Grauman, P.; Hurst, S.N.; McConkey, M.; Svinkina, T.; Heckl, D.; Comer, E.; Li, X.; et al. Lenalidomide Causes Selective Degradation of IKZF1 and IKZF3 in Multiple Myeloma Cells. Science 2014, 343, 301–305. [Google Scholar] [CrossRef]
- McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef] [PubMed]
- Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef]
- Kim, H.; Zheng, S.; Amini, S.S.; Virk, S.M.; Mikkelsen, T.; Brat, D.J.; Grimsby, J.; Sougnez, C.; Muller, F.; Hu, J.; et al. Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution. Genome Res. 2015, 25, 316–327. [Google Scholar] [CrossRef] [PubMed]
- Coombs, C.C.; Zehir, A.; Devlin, S.M.; Kishtagari, A.; Syed, A.; Jonsson, P.; Hyman, D.M.; Solit, D.B.; Robson, M.E.; Baselga, J.; et al. Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes. Cell Stem. Cell 2017, 21, 374–382 e4. [Google Scholar] [CrossRef]
- Lindsley, R.C.; Saber, W.; Mar, B.G.; Redd, R.; Wang, T.; Haagenson, M.D.; Grauman, P.V.; Hu, Z.-H.; Spellman, S.R.; Lee, S.J.; et al. Prognostic Mutations in Myelodysplastic Syndrome after Stem-Cell Transplantation. N. Engl. J. Med. 2017, 376, 536–547. [Google Scholar] [CrossRef] [PubMed]
- Baish, J.W.; Jain, R.K. Fractals and Cancer1. Cancer Res. 2000, 60, 3683–3688. [Google Scholar] [PubMed]
- Vakoc, B.J.; Lanning, R.M.; Tyrrell, J.A.; Padera, T.P.; Bartlett, L.A.; Stylianopoulos, T.; Munn, L.L.; Tearney, G.J.; Fukumura, D.; Jain, R.K.; et al. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat. Med. 2009, 15, 1219–1223. [Google Scholar] [CrossRef]
- Hobbs, S.K.; Monsky, W.L.; Yuan, F.; Roberts, W.G.; Griffith, L.; Torchilin, V.P.; Jain, R.K. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc. Natl. Acad. Sci. USA 1998, 95, 4607–4612. [Google Scholar] [CrossRef]
- Jain, R.K. Transport of molecules across tumor vasculature. Cancer Metastasis Rev. 1987, 6, 559–593. [Google Scholar] [CrossRef]
- Jain, R.K.; Stylianopoulos, T. Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol. 2010, 7, 653–664. [Google Scholar] [CrossRef]
- Endrich, B.; Reinhold, H.S.; Gross, J.F.; Intaglietta, M. Tissue Perfusion Inhomogeneity During Early Tumor Growth in Rats2. JNCI J. Natl. Cancer Inst. 1979, 62, 387–395. [Google Scholar] [CrossRef]
- Sanna, K.; Rofstad, E.K. Hypoxia-induced resistance to doxorubicin and methotrexate in human melanoma cell lines in vitro. Int. J. Cancer 1994, 58, 258–262. [Google Scholar] [CrossRef]
- Jain, R.K. Transport Phenomena in Tumors. In Advances in Chemical Engineering; Wei, J., Ed.; Academic Press: Cambridge, MA, USA, 1994; Volume 19, pp. 129–200. [Google Scholar]
- Kamoun, W.S.; Chae, S.-S.; Lacorre, D.A.; Tyrrell, J.A.; Mitre, M.; Gillissen, M.A.; Fukumura, D.; Jain, R.K.; Munn, L.L. Simultaneous measurement of RBC velocity, flux, hematocrit and shear rate in vascular networks. Nat. Methods 2010, 7, 655–660. [Google Scholar] [CrossRef] [PubMed]
- Yuan, F.; Dellian, M.; Fukumura, D.; Leunig, M.; Berk, D.A.; Torchilin, V.P.; Jain, R.K. Vascular Permeability in a Human Tumor Xenograft: Molecular Size Dependence and Cutoff Size1. Cancer Res. 1995, 55, 3752–3756. [Google Scholar] [PubMed]
- Yuan, F.; Salehi, H.A.; Boucher, Y.; Vasthare, U.S.; Tuma, R.F.; Jain, R.K. Vascular Permeability and Microcirculation of Gliomas and Mammary Carcinomas Transplanted in Rat and Mouse Cranial Windows1. Cancer Res. 1994, 54, 4564–4568. [Google Scholar]
- Hagendoorn, J.; Tong, R.; Fukumura, D.; Lin, Q.; Lobo, J.; Padera, T.P.; Xu, L.; Kucherlapati, R.; Jain, R.K. Onset of Abnormal Blood and Lymphatic Vessel Function and Interstitial Hypertension in Early Stages of Carcinogenesis. Cancer Res. 2006, 66, 3360–3364. [Google Scholar] [CrossRef] [PubMed]
- Padera, T.P.; Stoll, B.R.; Tooredman, J.B.; Capen, D.; Tomaso, E.D.; Jain, R.K. Cancer cells compress intratumour vessels. Nature 2004, 427, 695. [Google Scholar] [CrossRef]
- Boucher, Y.; Baxter, L.T.; Jain, R.K. Interstitial Pressure Gradients in Tissue-isolated and Subcutaneous Tumors: Implications for Therapy1. Cancer Res. 1990, 50, 4478–4484. [Google Scholar]
- Jain, R.K.; Baxter, L.T. Mechanisms of Heterogeneous Distribution of Monoclonal Antibodies and Other Macromolecules in Tumors: Significance of Elevated Interstitial Pressure1. Cancer Res. 1988, 48, 7022–7032. [Google Scholar] [PubMed]
- Jain, R.K.; Tong, R.T.; Munn, L.L. Effect of Vascular Normalization by Antiangiogenic Therapy on Interstitial Hypertension, Peritumor Edema, and Lymphatic Metastasis: Insights from a Mathematical Model. Cancer Res. 2007, 67, 2729–2735. [Google Scholar] [CrossRef]
- Baxter, L.T.; Jain, R.K. Transport of fluid and macromolecules in tumors. I. Role of interstitial pressure and convection. Microvasc. Res. 1989, 37, 77–104. [Google Scholar] [CrossRef]
- Baxter, L.T.; Jain, R.K. Transport of fluid and macromolecules in tumors. II. Role of heterogeneous perfusion and lymphatics. Microvasc. Res. 1990, 40, 246–263. [Google Scholar] [CrossRef]
- Jain, R.K. Delivery of Novel Therapeutic Agents in Tumors: Physiological Barriers and Strategies. JNCI J. Natl. Cancer Inst. 1990, 81, 570–576. [Google Scholar] [CrossRef] [PubMed]
- Spivak-Kroizman, T.R.; Hostetter, G.; Posner, R.; Aziz, M.; Hu, C.; Demeure, M.J.; Von Hoff, D.; Hingorani, S.R.; Palculict, T.B.; Izzo, J.; et al. Hypoxia Triggers Hedgehog-Mediated Tumor–Stromal Interactions in Pancreatic Cancer. Cancer Res. 2013, 73, 3235–3247. [Google Scholar] [CrossRef] [PubMed]
- Stylianopoulos, T.; Martin, J.D.; Chauhan, V.P.; Jain, S.R.; Diop-Frimpong, B.; Bardeesy, N.; Smith, B.L.; Ferrone, C.R.; Hornicek, F.J.; Boucher, Y.; et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 15101–15108. [Google Scholar] [CrossRef]
- Yamada, K.M.; Collins, J.W.; Cruz Walma, D.A.; Doyle, A.D.; Morales, S.G.; Lu, J.; Matsumoto, K.; Nazari, S.S.; Sekiguchi, R.; Shinsato, Y.; et al. Extracellular matrix dynamics in cell migration, invasion and tissue morphogenesis. Int. J. Exp. Pathol. 2019, 100, 144–152. [Google Scholar] [CrossRef]
- Bissell, M.J.; Hall, H.G.; Parry, G. How does the extracellular matrix direct gene expression? J. Theor. Biol. 1982, 99, 31–68. [Google Scholar] [CrossRef]
- Pearce, O.M.T.; Delaine-Smith, R.M.; Maniati, E.; Nichols, S.; Wang, J.; Böhm, S.; Rajeeve, V.; Ullah, D.; Chakravarty, P.; Jones, R.R.; et al. Deconstruction of a Metastatic Tumor Microenvironment Reveals a Common Matrix Response in Human Cancers. Cancer Discov. 2018, 8, 304–319. [Google Scholar] [CrossRef]
- Thorsson, V.; Gibbs, D.L.; Brown, S.D.; Wolf, D.; Bortone, D.S.; Ou Yang, T.-H.; Porta-Pardo, E.; Gao, G.F.; Plaisier, C.L.; Eddy, J.A.; et al. The Immune Landscape of Cancer. Immunity 2018, 48, 812–830. [Google Scholar] [CrossRef]
- Cox, T.R. The matrix in cancer. Nat. Rev. Cancer 2021, 21, 217–238. [Google Scholar] [CrossRef]
- Amatangelo, M.D.; Bassi, D.E.; Klein-Szanto, A.J.P.; Cukierman, E. Stroma-Derived Three-Dimensional Matrices Are Necessary and Sufficient to Promote Desmoplastic Differentiation of Normal Fibroblasts. Am. J. Pathol. 2005, 167, 475–488. [Google Scholar] [CrossRef] [PubMed]
- Piersma, B.; Hayward, M.-K.; Weaver, V.M. Fibrosis and cancer: A strained relationship. Biochim. Et Biophys. Acta (BBA) Rev. Cancer 2020, 1873, 188356. [Google Scholar] [CrossRef]
- Vitale, D.; Kumar Katakam, S.; Greve, B.; Jang, B.; Oh, E.-S.; Alaniz, L.; Götte, M. Proteoglycans and glycosaminoglycans as regulators of cancer stem cell function and therapeutic resistance. FEBS J. 2019, 286, 2870–2882. [Google Scholar] [CrossRef] [PubMed]
- Pires, A.; Greenshields-Watson, A.; Jones, E.; Smart, K.; Lauder, S.N.; Somerville, M.; Milutinovic, S.; Kendrick, H.; Hindley, J.P.; French, R.; et al. Immune Remodeling of the Extracellular Matrix Drives Loss of Cancer Stem Cells and Tumor Rejection. Cancer Immunol. Res. 2020, 8, 1520–1531. [Google Scholar] [CrossRef]
- Filipe, E.C.; Chitty, J.L.; Cox, T.R. Charting the unexplored extracellular matrix in cancer. Int. J. Exp. Pathol. 2018, 99, 58–76. [Google Scholar] [CrossRef] [PubMed]
- Nam, S.; Hu, K.H.; Butte, M.J.; Chaudhuri, O. Strain-enhanced stress relaxation impacts nonlinear elasticity in collagen gels. Proc. Natl. Acad. Sci. USA 2016, 113, 5492–5497. [Google Scholar] [CrossRef]
- Yue, X.; Nguyen, T.D.; Zellmer, V.; Zhang, S.; Zorlutuna, P. Stromal cell-laden 3D hydrogel microwell arrays as tumor microenvironment model for studying stiffness dependent stromal cell-cancer interactions. Biomaterials 2018, 170, 37–48. [Google Scholar] [CrossRef]
- Tian, C.; Clauser, K.R.; Öhlund, D.; Rickelt, S.; Huang, Y.; Gupta, M.; Mani, D.R.; Carr, S.A.; Tuveson, D.A.; Hynes, R.O. Proteomic analyses of ECM during pancreatic ductal adenocarcinoma progression reveal different contributions by tumor and stromal cells. Proc. Natl. Acad. Sci. USA 2019, 116, 19609–19618. [Google Scholar] [CrossRef]
- Kuninty, P.R.; Bansal, R.; De Geus, S.W.L.; Mardhian, D.F.; Schnittert, J.; van Baarlen, J.; Storm, G.; Bijlsma, M.F.; van Laarhoven, H.W.; Metselaar, J.M.; et al. ITGA5 inhibition in pancreatic stellate cells attenuates desmoplasia and potentiates efficacy of chemotherapy in pancreatic cancer. Sci. Adv. 2019, 5, eaax2770. [Google Scholar] [CrossRef]
- Below, C.R.; Kelly, J.; Brown, A.; Humphries, J.D.; Hutton, C.; Xu, J.; Lee, B.Y.; Cintas, C.; Zhang, X.; Hernandez-Gordillo, V.; et al. A microenvironment-inspired synthetic three-dimensional model for pancreatic ductal adenocarcinoma organoids. Nat. Mater. 2022, 21, 110–119. [Google Scholar] [CrossRef] [PubMed]
- Rice, A.J.; Cortes, E.; Lachowski, D.; Cheung, B.C.H.; Karim, S.A.; Morton, J.P.; del Río Hernández, A. Matrix stiffness induces epithelial–mesenchymal transition and promotes chemoresistance in pancreatic cancer cells. Oncogenesis 2017, 6, e352. [Google Scholar] [CrossRef]
- Hamidi, H.; Ivaska, J. Every step of the way: Integrins in cancer progression and metastasis. Nat. Rev. Cancer 2018, 18, 533–548. [Google Scholar] [CrossRef]
- Yang, X.H.; Flores, L.M.; Li, Q.; Zhou, P.; Xu, F.; Krop, I.E.; Hemler, M.E. Disruption of Laminin-Integrin-CD151-Focal Adhesion Kinase Axis Sensitizes Breast Cancer Cells to ErbB2 Antagonists. Cancer Res. 2010, 70, 2256–2263. [Google Scholar] [CrossRef] [PubMed]
- Pupa, S.M.; Giuffré, S.; Castiglioni, F.; Bertola, L.; Cantú, M.; Bongarzone, I.; Baldassari, P.; Mortarini, R.; Argraves, W.S.; Anichini, A.; et al. Regulation of Breast Cancer Response to Chemotherapy by Fibulin-1. Cancer Res. 2007, 67, 4271–4277. [Google Scholar] [CrossRef] [PubMed]
- Hirata, E.; Girotti, M.R.; Viros, A.; Hooper, S.; Spencer-Dene, B.; Matsuda, M.; Larkin, J.; Marais, R.; Sahai, E. Intravital Imaging Reveals How BRAF Inhibition Generates Drug-Tolerant Microenvironments with High Integrin β1/FAK Signaling. Cancer Cell 2015, 27, 574–588. [Google Scholar] [CrossRef] [PubMed]
- Crawford, Y.; Kasman, I.; Yu, L.; Zhong, C.; Wu, X.; Modrusan, Z.; Kaminker, J.; Ferrara, N. PDGF-C Mediates the Angiogenic and Tumorigenic Properties of Fibroblasts Associated with Tumors Refractory to Anti-VEGF Treatment. Cancer Cell 2009, 15, 21–34. [Google Scholar] [CrossRef]
- O’Brown, N.M.; Pfau, S.J.; Gu, C. Bridging barriers: A comparative look at the blood-brain barrier across organisms. Genes Dev. 2018, 32, 466–478. [Google Scholar] [CrossRef]
- Hendricks, B.K.; Cohen-Gadol, A.A.; Miller, J.C. Novel delivery methods bypassing the blood-brain and blood-tumor barriers. Neurosurg. Focus FOC 2015, 38, E10. [Google Scholar] [CrossRef]
- Abbott, N.J.; Rönnbäck, L.; Hansson, E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 2006, 7, 41–53. [Google Scholar] [CrossRef]
- Giepmans, B.N.G.; van Ijzendoorn, S.C.D. Epithelial cell–cell junctions and plasma membrane domains. Biochim. Et Biophys. Acta (BBA) Biomembr. 2009, 1788, 820–831. [Google Scholar] [CrossRef]
- Agarwal, S.; Sane, R.; Gallardo, J.L.; Ohlfest, J.R.; Elmquist, W.F. Distribution of Gefitinib to the Brain Is Limited by P-glycoprotein (ABCB1) and Breast Cancer Resistance Protein (ABCG2)-Mediated Active Efflux. J. Pharmacol. Exp. Ther. 2010, 334, 147–155. [Google Scholar] [CrossRef]
- Demeule, M.; Régina, A.; Jodoin, J.; Laplante, A.; Dagenais, C.; Berthelet, F.; Moghrabi, A.; Béliveau, R. Drug transport to the brain: Key roles for the efflux pump P-glycoprotein in the blood–brain barrier. Vasc. Pharmacol. 2002, 38, 339–348. [Google Scholar] [CrossRef]
- Wang, D.; Wang, C.; Wang, L.; Chen, Y. A comprehensive review in improving delivery of small-molecule chemotherapeutic agents overcoming the blood-brain/brain tumor barriers for glioblastoma treatment. Drug Deliv. 2019, 26, 551–565. [Google Scholar] [CrossRef] [PubMed]
- Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
- Monsky, W.L.; Carreira, C.M.; Tsuzuki, Y.; Gohongi, T.; Fukumura, D.; Jain, R.K. Role of Host Microenvironment in Angiogenesis and Microvascular Functions in Human Breast Cancer Xenografts: Mammary Fat Pad versus Cranial Tumors1. Clin. Cancer Res. 2002, 8, 1008–1013. [Google Scholar] [PubMed]
- Pitz, M.W.; Desai, A.; Grossman, S.A.; Blakeley, J.O. Tissue concentration of systemically administered antineoplastic agents in human brain tumors. J. Neuro-Oncol. 2011, 104, 629–638. [Google Scholar] [CrossRef] [PubMed]
- Deeken, J.F.; Löscher, W. The Blood-Brain Barrier and Cancer: Transporters, Treatment, and Trojan Horses. Clin. Cancer Res. 2007, 13, 1663–1674. [Google Scholar] [CrossRef]
- Sarkaria, J.N.; Hu, L.S.; Parney, I.F.; Pafundi, D.H.; Brinkmann, D.H.; Laack, N.N.; Giannini, C.; Burns, T.C.; Kizilbash, S.H.; Laramy, J.K.; et al. Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro-Oncology 2017, 20, 184–191. [Google Scholar] [CrossRef]
- Gerlinger, M.; Rowan, A.J.; Horswell, S.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; Tarpey, P.; et al. Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef] [PubMed]
- Ha, G.; Roth, A.; Khattra, J.; Ho, J.; Yap, D.; Prentice, L.M.; Melnyk, N.; McPherson, A.; Bashashati, A.; Laks, E.; et al. TITAN: Inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 2014, 24, 1881–1893. [Google Scholar] [CrossRef]
- Roth, A.; Khattra, J.; Yap, D.; Wan, A.; Laks, E.; Biele, J.; Ha, G.; Aparicio, S.; Bouchard-Côté, A.; Shah, S.P. PyClone: Statistical inference of clonal population structure in cancer. Nat. Methods 2014, 11, 396–398. [Google Scholar] [CrossRef]
- Van Loo, P.; Nordgard, S.H.; Lingjærde, O.C.; Russnes, H.G.; Rye, I.H.; Sun, W.; Weigman, V.J.; Marynen, P.; Zetterberg, A.; Naume, B.; et al. Allele-specific copy number analysis of tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 16910–16915. [Google Scholar] [CrossRef]
- Haber, D.A.; Velculescu, V.E. Blood-Based Analyses of Cancer: Circulating Tumor Cells and Circulating Tumor DNA. Cancer Discov. 2014, 4, 650–661. [Google Scholar] [CrossRef] [PubMed]
- Kotani, D.; Oki, E.; Nakamura, Y.; Yukami, H.; Mishima, S.; Bando, H.; Shirasu, H.; Yamazaki, K.; Watanabe, J.; Kotaka, M.; et al. Molecular residual disease and efficacy of adjuvant chemotherapy in patients with colorectal cancer. Nat. Med. 2023, 29, 127–134. [Google Scholar] [CrossRef] [PubMed]
- Misale, S.; Arena, S.; Lamba, S.; Siravegna, G.; Lallo, A.; Hobor, S.; Russo, M.; Buscarino, M.; Lazzari, L.; Sartore-Bianchi, A.; et al. Blockade of EGFR and MEK Intercepts Heterogeneous Mechanisms of Acquired Resistance to Anti-EGFR Therapies in Colorectal Cancer. Sci. Transl. Med. 2014, 6, 224ra226. [Google Scholar] [CrossRef]
- Bozic, I.; Nowak, M.A. Timing and heterogeneity of mutations associated with drug resistance in metastatic cancers. Proc. Natl. Acad. Sci. USA 2014, 111, 15964–15968. [Google Scholar] [CrossRef]
- Palmer, C.D.; Rappaport, A.R.; Davis, M.J.; Hart, M.G.; Scallan, C.D.; Hong, S.-J.; Gitlin, L.; Kraemer, L.D.; Kounlavouth, S.; Yang, A.; et al. Individualized, heterologous chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced metastatic solid tumors: Phase 1 trial interim results. Nat. Med. 2022, 28, 1619–1629. [Google Scholar] [CrossRef]
- Rappaport, A.R.; Kyi, C.; Lane, M.; Hart, M.G.; Johnson, M.L.; Henick, B.S.; Liao, C.-Y.; Mahipal, A.; Shergill, A.; Spira, A.I.; et al. A shared neoantigen vaccine combined with immune checkpoint blockade for advanced metastatic solid tumors: Phase 1 trial interim results. Nat. Med. 2024, 30, 1013–1022. [Google Scholar] [CrossRef]
- Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H.; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef] [PubMed]
- Crowley, E.; Di Nicolantonio, F.; Loupakis, F.; Bardelli, A. Liquid biopsy: Monitoring cancer-genetics in the blood. Nat. Rev. Clin. Oncol. 2013, 10, 472–484. [Google Scholar] [CrossRef]
- Wagle, N.; Emery, C.; Berger, M.F.; Davis, M.J.; Sawyer, A.; Pochanard, P.; Kehoe, S.M.; Johannessen, C.M.; MacConaill, L.E.; Hahn, W.C.; et al. Dissecting Therapeutic Resistance to RAF Inhibition in Melanoma by Tumor Genomic Profiling. J. Clin. Oncol. 2011, 29, 3085–3096. [Google Scholar] [CrossRef]
- Gorre, M.E.; Mohammed, M.; Ellwood, K.; Hsu, N.; Paquette, R.; Rao, P.N.; Sawyers, C.L. Clinical Resistance to STI-571 Cancer Therapy Caused by BCR-ABL Gene Mutation or Amplification. Science 2001, 293, 876–880. [Google Scholar] [CrossRef]
- Diaz, L.A., Jr.; Williams, R.T.; Wu, J.; Kinde, I.; Hecht, J.R.; Berlin, J.; Allen, B.; Bozic, I.; Reiter, J.G.; Nowak, M.A.; et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature 2012, 486, 537–540. [Google Scholar] [CrossRef] [PubMed]
- Misale, S.; Yaeger, R.; Hobor, S.; Scala, E.; Janakiraman, M.; Liska, D.; Valtorta, E.; Schiavo, R.; Buscarino, M.; Siravegna, G.; et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature 2012, 486, 532–536. [Google Scholar] [CrossRef] [PubMed]
- Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and Histological Evolution of Lung Cancers Acquiring Resistance to EGFR Inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef] [PubMed]
- Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.-K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef]
- Choi, Y.L.; Soda, M.; Yamashita, Y.; Ueno, T.; Takashima, J.; Nakajima, T.; Yatabe, Y.; Takeuchi, K.; Hamada, T.; Haruta, H.; et al. EML4-ALK Mutations in Lung Cancer That Confer Resistance to ALK Inhibitors. N. Engl. J. Med. 2010, 363, 1734–1739. [Google Scholar] [CrossRef]
- Yun, C.-H.; Mengwasser, K.E.; Toms, A.V.; Woo, M.S.; Greulich, H.; Wong, K.-K.; Meyerson, M.; Eck, M.J. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. USA 2008, 105, 2070–2075. [Google Scholar] [CrossRef]
- Luo, L.; Parrish, C.A.; Nevins, N.; McNulty, D.E.; Chaudhari, A.M.; Carson, J.D.; Sudakin, V.; Shaw, A.N.; Lehr, R.; Zhao, H.; et al. ATP-competitive inhibitors of the mitotic kinesin KSP that function via an allosteric mechanism. Nat. Chem. Biol. 2007, 3, 722–726. [Google Scholar] [CrossRef]
- Diehl, F.; Schmidt, K.; Choti, M.A.; Romans, K.; Goodman, S.; Li, M.; Thornton, K.; Agrawal, N.; Sokoll, L.; Szabo, S.A.; et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008, 14, 985–990. [Google Scholar] [CrossRef]
- Frattini, M.; Gallino, G.; Signoroni, S.; Balestra, D.; Lusa, L.; Battaglia, L.; Sozzi, G.; Bertario, L.; Leo, E.; Pilotti, S.; et al. Quantitative and qualitative characterization of plasma DNA identifies primary and recurrent colorectal cancer. Cancer Lett. 2008, 263, 170–181. [Google Scholar] [CrossRef]
- Leary, R.J.; Sausen, M.; Kinde, I.; Papadopoulos, N.; Carpten, J.D.; Craig, D.; O’Shaughnessy, J.; Kinzler, K.W.; Parmigiani, G.; Vogelstein, B.; et al. Detection of Chromosomal Alterations in the Circulation of Cancer Patients with Whole-Genome Sequencing. Sci. Transl. Med. 2012, 4, 162ra154. [Google Scholar] [CrossRef]
- Forshew, T.; Murtaza, M.; Parkinson, C.; Gale, D.; Tsui, D.W.Y.; Kaper, F.; Dawson, S.-J.; Piskorz, A.M.; Jimenez-Linan, M.; Bentley, D.; et al. Noninvasive Identification and Monitoring of Cancer Mutations by Targeted Deep Sequencing of Plasma DNA. Sci. Transl. Med. 2012, 4, 136ra168. [Google Scholar] [CrossRef] [PubMed]
- Kuhlmann, J.D.; Schwarzenbach, H.; Wimberger, P.; Poetsch, M.; Kimmig, R.; Kasimir-Bauer, S. LOH at 6q and 10q in fractionated circulating DNA of ovarian cancer patients is predictive for tumor cell spread and overall survival. BMC Cancer 2012, 12, 325. [Google Scholar] [CrossRef] [PubMed]
- Chan, K.A.; Jiang, P.; Zheng, Y.W.; Liao, G.J.; Sun, H.; Wong, J.; Siu, S.S.N.; Chan, W.C.; Chan, S.L.; Chan, A.T.; et al. Cancer Genome Scanning in Plasma: Detection of Tumor-Associated Copy Number Aberrations, Single-Nucleotide Variants, and Tumoral Heterogeneity by Massively Parallel Sequencing. Clin. Chem. 2013, 59, 211–224. [Google Scholar] [CrossRef] [PubMed]
- Nygaard, A.D.; Garm Spindler, K.-L.; Pallisgaard, N.; Andersen, R.F.; Jakobsen, A. The prognostic value of KRAS mutated plasma DNA in advanced non-small cell lung cancer. Lung Cancer 2013, 79, 312–317. [Google Scholar] [CrossRef]
- Santarpia, L.; Qi, Y.; Stemke-Hale, K.; Wang, B.; Young, E.J.; Booser, D.J.; Holmes, F.A.; O’Shaughnessy, J.; Hellerstedt, B.; Pippen, J.; et al. Mutation profiling identifies numerous rare drug targets and distinct mutation patterns in different clinical subtypes of breast cancers. Breast Cancer Res. Treat. 2012, 134, 333–343. [Google Scholar] [CrossRef]
- Dawson, S.-J.; Tsui, D.W.Y.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.-F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef]
- Higgins, M.J.; Jelovac, D.; Barnathan, E.; Blair, B.; Slater, S.; Powers, P.; Zorzi, J.; Jeter, S.C.; Oliver, G.R.; Fetting, J.; et al. Detection of Tumor PIK3CA Status in Metastatic Breast Cancer Using Peripheral Blood. Clin. Cancer Res. 2012, 18, 3462–3469. [Google Scholar] [CrossRef]
- Murtaza, M.; Dawson, S.-J.; Tsui, D.W.Y.; Gale, D.; Forshew, T.; Piskorz, A.M.; Parkinson, C.; Chin, S.-F.; Kingsbury, Z.; Wong, A.S.C.; et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 2013, 497, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Isakoff, S.J.; Engelman, J.A.; Irie, H.Y.; Luo, J.; Brachmann, S.M.; Pearline, R.V.; Cantley, L.C.; Brugge, J.S. Breast Cancer–Associated PIK3CA Mutations Are Oncogenic in Mammary Epithelial Cells. Cancer Res. 2005, 65, 10992–11000. [Google Scholar] [CrossRef]
- Kitai, H.; Choi, P.H.; Yang, Y.C.; Boyer, J.A.; Whaley, A.; Pancholi, P.; Thant, C.; Reiter, J.; Chen, K.; Markov, V.; et al. Combined inhibition of KRASG12C and mTORC1 kinase is synergistic in non-small cell lung cancer. Nat. Commun. 2024, 15, 6076. [Google Scholar] [CrossRef]
- Shi, H.; Moriceau, G.; Kong, X.; Lee, M.-K.; Lee, H.; Koya, R.C.; Ng, C.; Chodon, T.; Scolyer, R.A.; Dahlman, K.B.; et al. Melanoma whole-exome sequencing identifies V600EB-RAF amplification-mediated acquired B-RAF inhibitor resistance. Nat. Commun. 2012, 3, 724. [Google Scholar] [CrossRef] [PubMed]
- Kantarjian, H.M.; Larson, R.A.; Guilhot, F.; O’Brien, S.G.; Mone, M.; Rudoltz, M.; Krahnke, T.; Cortes, J.; Druker, B.J.; International Randomized Study of Interferon and STI571 (IRIS) Investigators. Efficacy of imatinib dose escalation in patients with chronic myeloid leukemia in chronic phase. Cancer 2009, 115, 551–560. [Google Scholar] [CrossRef] [PubMed]
- Kantarjian, H.; Pasquini, R.; Lévy, V.; Jootar, S.; Holowiecki, J.; Hamerschlak, N.; Hughes, T.; Bleickardt, E.; Dejardin, D.; Cortes, J.; et al. Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia resistant to imatinib at a dose of 400 to 600 milligrams daily. Cancer 2009, 115, 4136–4147. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, S.; Boggon, T.J.; Dayaram, T.; Jänne, P.A.; Kocher, O.; Meyerson, M.; Johnson, B.E.; Eck, M.J.; Tenen, D.G.; Halmos, B. EGFR Mutation and Resistance of Non–Small-Cell Lung Cancer to Gefitinib. N. Engl. J. Med. 2005, 352, 786–792. [Google Scholar] [CrossRef]
- Shah, N.P.; Nicoll, J.M.; Nagar, B.; Gorre, M.E.; Paquette, R.L.; Kuriyan, J.; Sawyers, C.L. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2002, 2, 117–125. [Google Scholar] [CrossRef]
- Ogino, A.; Kitao, H.; Hirano, S.; Uchida, A.; Ishiai, M.; Kozuki, T.; Takigawa, N.; Takata, M.; Kiura, K.; Tanimoto, M. Emergence of Epidermal Growth Factor Receptor T790M Mutation during Chronic Exposure to Gefitinib in a Non–Small Cell Lung Cancer Cell Line. Cancer Res. 2007, 67, 7807–7814. [Google Scholar] [CrossRef]
- Deininger, M.; Buchdunger, E.; Druker, B.J. The development of imatinib as a therapeutic agent for chronic myeloid leukemia. Blood 2005, 105, 2640–2653. [Google Scholar] [CrossRef]
- Linardou, H.; Dahabreh, I.J.; Bafaloukos, D.; Kosmidis, P.; Murray, S. Somatic EGFR mutations and efficacy of tyrosine kinase inhibitors in NSCLC. Nat. Rev. Clin. Oncol. 2009, 6, 352–366. [Google Scholar] [CrossRef]
- Carter, T.A.; Wodicka, L.M.; Shah, N.P.; Velasco, A.M.; Fabian, M.A.; Treiber, D.K.; Milanov, Z.V.; Atteridge, C.E.; Biggs, W.H.; Edeen, P.T.; et al. Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases. Proc. Natl. Acad. Sci. USA 2005, 102, 11011–11016. [Google Scholar] [CrossRef]
- Bozic, I.; Reiter, J.G.; Allen, B.; Antal, T.; Chatterjee, K.; Shah, P.; Moon, Y.S.; Yaqubie, A.; Kelly, N.; Le, D.T.; et al. Evolutionary dynamics of cancer in response to targeted combination therapy. eLife 2013, 2, e00747. [Google Scholar] [CrossRef]
- Komarova, N.L.; Katouli, A.A.; Wodarz, D. Combination of Two but Not Three Current Targeted Drugs Can Improve Therapy of Chronic Myeloid Leukemia. PLoS ONE 2009, 4, e4423. [Google Scholar] [CrossRef] [PubMed]
- Katouli, A.A.; Komarova, N.L. The Worst Drug Rule Revisited: Mathematical Modeling of Cyclic Cancer Treatments. Bull. Math. Biol. 2011, 73, 549–584. [Google Scholar] [CrossRef] [PubMed]
- McGranahan, N.; Furness, A.J.S.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469. [Google Scholar] [CrossRef] [PubMed]
- Folkman, J. Successful Treatment of an Angiogenic Disease. N. Engl. J. Med. 1989, 320, 1211–1212. [Google Scholar] [CrossRef]
- Boehm, T.; Folkman, J.; Browder, T.; O’Reilly, M.S. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 1997, 390, 404–407. [Google Scholar] [CrossRef]
- Jain, R.K. Normalizing tumor vasculature with anti-angiogenic therapy: A new paradigm for combination therapy. Nat. Med. 2001, 7, 987–989. [Google Scholar] [CrossRef]
- Leung, D.W.; Cachianes, G.; Kuang, W.-J.; Goeddel, D.V.; Ferrara, N. Vascular Endothelial Growth Factor Is a Secreted Angiogenic Mitogen. Science 1989, 246, 1306–1309. [Google Scholar] [CrossRef]
- Kerr, D.J. Targeting angiogenesis in cancer: Clinical development of bevacizumab. Nat. Clin. Pract. Oncol. 2004, 1, 39–43. [Google Scholar] [CrossRef]
- Jones, L.H. Medicinal Chemical Biology. In Burger’s Medicinal Chemistry and Drug Discovery; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2021; pp. 1–37. [Google Scholar] [CrossRef]
- Miller, K.; Wang, M.; Gralow, J.; Dickler, M.; Cobleigh, M.; Perez, E.A.; Shenkier, T.; Cella, D.; Davidson, N.E. Paclitaxel plus Bevacizumab versus Paclitaxel Alone for Metastatic Breast Cancer. N. Engl. J. Med. 2007, 357, 2666–2676. [Google Scholar] [CrossRef]
- Reck, M.; Pawel, J.V.; Zatloukal, P.; Ramlau, R.; Gorbounova, V.; Hirsh, V.; Leighl, N.; Mezger, J.; Archer, V.; Moore, N.; et al. Phase III Trial of Cisplatin Plus Gemcitabine with Either Placebo or Bevacizumab as First-Line Therapy for Nonsquamous Non–Small-Cell Lung Cancer: AVAiL. J. Clin. Oncol. 2009, 27, 1227–1234. [Google Scholar] [CrossRef]
- Tolaney, S.M.; Boucher, Y.; Duda, D.G.; Martin, J.D.; Seano, G.; Ancukiewicz, M.; Barry, W.T.; Goel, S.; Lahdenrata, J.; Isakoff, S.J.; et al. Role of vascular density and normalization in response to neoadjuvant bevacizumab and chemotherapy in breast cancer patients. Proc. Natl. Acad. Sci. USA 2015, 112, 14325–14330. [Google Scholar] [CrossRef] [PubMed]
- Winkler, F.; Kozin, S.V.; Tong, R.T.; Chae, S.-S.; Booth, M.F.; Garkavtsev, I.; Xu, L.; Hicklin, D.J.; Fukumura, D.; di Tomaso, E.; et al. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiation: Role of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 2004, 6, 553–563. [Google Scholar] [CrossRef]
- Tong, R.T.; Boucher, Y.; Kozin, S.V.; Winkler, F.; Hicklin, D.J.; Jain, R.K. Vascular Normalization by Vascular Endothelial Growth Factor Receptor 2 Blockade Induces a Pressure Gradient across the Vasculature and Improves Drug Penetration in Tumors. Cancer Res. 2004, 64, 3731–3736. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Q.; Gallo, J.M. Differential effect of sunitinib on the distribution of temozolomide in an orthotopic glioma model. Neuro-Oncology 2009, 11, 301–310. [Google Scholar] [CrossRef]
- Zhou, Q.; Guo, P.; Gallo, J.M. Impact of Angiogenesis Inhibition by Sunitinib on Tumor Distribution of Temozolomide. Clin. Cancer Res. 2008, 14, 1540–1549. [Google Scholar] [CrossRef]
- Jayson, G.C.; Kerbel, R.; Ellis, L.M.; Harris, A.L. Antiangiogenic therapy in oncology: Current status and future directions. Lancet 2016, 388, 518–529. [Google Scholar] [CrossRef] [PubMed]
- Xian, X.; Hakansson, J.; Stahlberg, A.; Lindblom, P.; Betsholtz, C.; Gerhardt, H.; Semb, H. Pericytes limit tumor cell metastasis. J. Clin. Investig. 2006, 116, 642–651. [Google Scholar] [CrossRef]
- Nasarre, P.; Thomas, M.; Kruse, K.; Helfrich, I.; Wolter, V.; Deppermann, C.; Schadendorf, D.; Thurston, G.; Fiedler, U.; Augustin, H.G. Host-Derived Angiopoietin-2 Affects Early Stages of Tumor Development and Vessel Maturation but Is Dispensable for Later Stages of Tumor Growth. Cancer Res. 2009, 69, 1324–1333. [Google Scholar] [CrossRef]
- Roos-Mattila, M.; Kaprio, T.; Mustonen, H.; Hagström, J.; Saharinen, P.; Haglund, C.; Seppänen, H. The possible dual role of Ang-2 in the prognosis of pancreatic cancer. Sci. Rep. 2023, 13, 18725. [Google Scholar] [CrossRef]
- Holopainen, T.; Saharinen, P.; D’Amico, G.; Lampinen, A.; Eklund, L.; Sormunen, R.; Anisimov, A.; Zarkada, G.; Lohela, M.; Heloterä, H.; et al. Effects of Angiopoietin-2-Blocking Antibody on Endothelial Cell–Cell Junctions and Lung Metastasis. JNCI J. Natl. Cancer Inst. 2012, 104, 461–475. [Google Scholar] [CrossRef]
- Monk, B.J.; Poveda, A.; Vergote, I.; Raspagliesi, F.; Fujiwara, K.; Bae, D.-S.; Oaknin, A.; Ray-Coquard, I.; Provencher, D.M.; Karlan, B.Y.; et al. Final results of a phase 3 study of trebananib plus weekly paclitaxel in recurrent ovarian cancer (TRINOVA-1): Long-term survival, impact of ascites, and progression-free survival-2. Gynecol. Oncol. 2016, 143, 27–34. [Google Scholar] [CrossRef] [PubMed]
- Goel, S.; Gupta, N.; Walcott, B.P.; Snuderl, M.; Kesler, C.T.; Kirkpatrick, N.D.; Heishi, T.; Huang, Y.; Martin, J.D.; Ager, E.; et al. Effects of Vascular-Endothelial Protein Tyrosine Phosphatase Inhibition on Breast Cancer Vasculature and Metastatic Progression. JNCI J. Natl. Cancer Inst. 2013, 105, 1188–1201. [Google Scholar] [CrossRef]
- Chakroborty, D.; Sarkar, C.; Yu, H.; Wang, J.; Liu, Z.; Dasgupta, P.S.; Basu, S. Dopamine stabilizes tumor blood vessels by up-regulating angiopoietin 1 expression in pericytes and Krüppel-like factor-2 expression in tumor endothelial cells. Proc. Natl. Acad. Sci. USA 2011, 108, 20730–20735. [Google Scholar] [CrossRef] [PubMed]
- Maharjan, S.; Kim, K.; Agrawal, V.; Choi, H.-J.; Kim, N.-J.; Kim, Y.-M.; Suh, Y.-G.; Kwon, Y.-G. Sac-1004, a novel vascular leakage blocker, enhances endothelial barrier through the cAMP/Rac/cortactin pathway. Biochem. Biophys. Res. Commun. 2013, 435, 420–427. [Google Scholar] [CrossRef]
- Agrawal, V.; Maharjan, S.; Kim, K.; Kim, N.J.; Son, J.; Lee, K.; Choi, H.J.; Rho, S.S.; Ahn, S.; Won, M.H.; et al. Direct endothelial junction restoration results in significant tumor vascular normalization and metastasis inhibition in mice. Oncotarget 2014, 5, 2761–2777. [Google Scholar] [CrossRef]
- Griffon-Etienne, G.E.; Boucher, Y.; Brekken, C.; Suit, H.D.; Jain, R.K. Taxane-induced Apoptosis Decompresses Blood Vessels and Lowers Interstitial Fluid Pressure in Solid Tumors: Clinical Implications1. Cancer Res. 1999, 59, 3776–3782. [Google Scholar] [PubMed]
- He, Y.; Kozaki, K.-i.; Karpanen, T.; Koshikawa, K.; Yla-Herttuala, S.; Takahashi, T.; Alitalo, K. Suppression of Tumor Lymphangiogenesis and Lymph Node Metastasis by Blocking Vascular Endothelial Growth Factor Receptor 3 Signaling. JNCI J. Natl. Cancer Inst. 2002, 94, 819–825. [Google Scholar] [CrossRef]
- Krishnan, J.; Kirkin, V.; Steffen, A.; Hegen, M.; Weih, D.; Tomarev, S.; Wilting, J.R.; Sleeman, J.P. Differential in Vivo and in Vitro Expression of Vascular Endothelial Growth Factor (VEGF)-C and VEGF-D in Tumors and Its Relationship to Lymphatic Metastasis in Immunocompetent Rats1. Cancer Res. 2003, 63, 713–722. [Google Scholar]
- Laakkonen, P.; Waltari, M.; Holopainen, T.; Takahashi, T.; Pytowski, B.; Steiner, P.; Hicklin, D.; Persaud, K.; Tonra, J.R.; Witte, L.; et al. Vascular Endothelial Growth Factor Receptor 3 Is Involved in Tumor Angiogenesis and Growth. Cancer Res. 2007, 67, 593–599. [Google Scholar] [CrossRef]
- Roberts, N.; Kloos, B.; Cassella, M.; Podgrabinska, S.; Persaud, K.; Wu, Y.; Pytowski, B.; Skobe, M. Inhibition of VEGFR-3 Activation with the Antagonistic Antibody More Potently Suppresses Lymph Node and Distant Metastases than Inactivation of VEGFR-2. Cancer Res. 2006, 66, 2650–2657. [Google Scholar] [CrossRef]
- Shimizu, K.; Kubo, H.; Yamaguchi, K.; Kawashima, K.; Ueda, Y.; Matsuo, K.; Awane, M.; Shimahara, Y.; Takabayashi, A.; Yamaoka, Y.; et al. Suppression of VEGFR-3 signaling inhibits lymph node metastasis in gastric cancer. Cancer Sci. 2004, 95, 328–333. [Google Scholar] [CrossRef] [PubMed]
- Fankhauser, M.; Broggi, M.A.S.; Potin, L.; Bordry, N.; Jeanbart, L.; Lund, A.W.; Da Costa, E.; Hauert, S.; Rincon-Restrepo, M.; Tremblay, C.; et al. Tumor lymphangiogenesis promotes T cell infiltration and potentiates immunotherapy in melanoma. Sci. Transl. Med. 2017, 9, eaal4712. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef]
- Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.-L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef]
- McKee, T.D.; Grandi, P.; Mok, W.; Alexandrakis, G.; Insin, N.; Zimmer, J.P.; Bawendi, M.G.; Boucher, Y.; Breakefield, X.O.; Jain, R.K. Degradation of Fibrillar Collagen in a Human Melanoma Xenograft Improves the Efficacy of an Oncolytic Herpes Simplex Virus Vector. Cancer Res. 2006, 66, 2509–2513. [Google Scholar] [CrossRef]
- Mok, W.; Boucher, Y.; Jain, R.K. Matrix Metalloproteinases-1 and -8 Improve the Distribution and Efficacy of an Oncolytic Virus. Cancer Res. 2007, 67, 10664–10668. [Google Scholar] [CrossRef]
- Perentes, J.Y.; McKee, T.D.; Ley, C.D.; Mathiew, H.; Dawson, M.; Padera, T.P.; Munn, L.L.; Jain, R.K.; Boucher, Y. In vivo imaging of extracellular matrix remodeling by tumor-associated fibroblasts. Nat. Methods 2009, 6, 143–145. [Google Scholar] [CrossRef] [PubMed]
- Nakai, Y.; Isayama, H.; Ijichi, H.; Sasaki, T.; Sasahira, N.; Hirano, K.; Kogure, H.; Kawakubo, K.; Yagioka, H.; Yashima, Y.; et al. Inhibition of renin–angiotensin system affects prognosis of advanced pancreatic cancer receiving gemcitabine. Br. J. Cancer 2010, 103, 1644–1648. [Google Scholar] [CrossRef]
- Wilop, S.; von Hobe, S.; Crysandt, M.; Esser, A.; Osieka, R.; Jost, E. Impact of angiotensin I converting enzyme inhibitors and angiotensin II type 1 receptor blockers on survival in patients with advanced non-small-cell lung cancer undergoing first-line platinum-based chemotherapy. J. Cancer Res. Clin. Oncol. 2009, 135, 1429–1435. [Google Scholar] [CrossRef]
- Keizman, D.; Huang, P.; Eisenberger, M.A.; Pili, R.; Kim, J.J.; Antonarakis, E.S.; Hammers, H.; Carducci, M.A. Angiotensin system inhibitors and outcome of sunitinib treatment in patients with metastatic renal cell carcinoma: A retrospective examination. Eur. J. Cancer 2011, 47, 1955–1961. [Google Scholar] [CrossRef]
- Liu, J.; Liao, S.; Diop-Frimpong, B.; Chen, W.; Goel, S.; Naxerova, K.; Ancukiewicz, M.; Boucher, Y.; Jain, R.K.; Xu, L. TGF-β blockade improves the distribution and efficacy of therapeutics in breast carcinoma by normalizing the tumor stroma. Proc. Natl. Acad. Sci. USA 2012, 109, 16618–16623. [Google Scholar] [CrossRef] [PubMed]
- Liao, S.; Liu, J.; Lin, P.; Shi, T.; Jain, R.K.; Xu, L. TGF-β Blockade Controls Ascites by Preventing Abnormalization of Lymphatic Vessels in Orthotopic Human Ovarian Carcinoma Models. Clin. Cancer Res. 2011, 17, 1415–1424. [Google Scholar] [CrossRef] [PubMed]
- Steeg, P.S.; Camphausen, K.A.; Smith, Q.R. Brain metastases as preventive and therapeutic targets. Nat. Rev. Cancer 2011, 11, 352–363. [Google Scholar] [CrossRef] [PubMed]
- Satcher, R.L.; Zhang, X.H.F. Evolving cancer–niche interactions and therapeutic targets during bone metastasis. Nat. Rev. Cancer 2022, 22, 85–101. [Google Scholar] [CrossRef]
- Lajoie, J.M.; Shusta, E.V. Targeting Receptor-Mediated Transport for Delivery of Biologics Across the Blood-Brain Barrier. Annu. Rev. Pharmacol. Toxicol. 2015, 55, 613–631. [Google Scholar] [CrossRef]
- Zhang, F.; Xu, C.L.; Liu, C.M. Drug delivery strategies to enhance the permeability of the blood-brain barrier for treatment of glioma. Drug Des. Devel. Ther. 2015, 9, 2089–2100. [Google Scholar] [CrossRef]
- de Vries, N.A.; Zhao, J.; Kroon, E.; Buckle, T.; Beijnen, J.H.; van Tellingen, O. P-Glycoprotein and Breast Cancer Resistance Protein: Two Dominant Transporters Working Together in Limiting the Brain Penetration of Topotecan. Clin. Cancer Res. 2007, 13, 6440–6449. [Google Scholar] [CrossRef]
- Szakács, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar] [CrossRef]
- Kusuhara, H.; Sugiyama, Y. ATP-binding cassette, subfamily G (ABCG family). Pflügers Arch. Eur. J. Physiol. 2007, 453, 735–744. [Google Scholar] [CrossRef]
- Lin, F.; de Gooijer, M.C.; Roig, E.M.; Buil, L.C.M.; Christner, S.M.; Beumer, J.H.; Würdinger, T.; Beijnen, J.H.; van Tellingen, O. ABCB1, ABCG2, and PTEN Determine the Response of Glioblastoma to Temozolomide and ABT-888 Therapy. Clin. Cancer Res. 2014, 20, 2703–2713. [Google Scholar] [CrossRef]
- Kemper, E.M.; Boogerd, W.; Thuis, I.; Beijnen, J.H.; van Tellingen, O. Modulation of the blood–brain barrier in oncology: Therapeutic opportunities for the treatment of brain tumours? Cancer Treat. Rev. 2004, 30, 415–423. [Google Scholar] [CrossRef] [PubMed]
- Arvanitis, C.D.; Askoxylakis, V.; Guo, Y.; Datta, M.; Kloepper, J.; Ferraro, G.B.; Bernabeu, M.O.; Fukumura, D.; McDannold, N.; Jain, R.K. Mechanisms of enhanced drug delivery in brain metastases with focused ultrasound-induced blood–tumor barrier disruption. Proc. Natl. Acad. Sci. USA 2018, 115, E8717–E8726. [Google Scholar] [CrossRef] [PubMed]
- VanBavel, E. Effects of shear stress on endothelial cells: Possible relevance for ultrasound applications. Prog. Biophys. Mol. Biol. 2007, 93, 374–383. [Google Scholar] [CrossRef]
- Jalali, S.; Huang, Y.; Dumont, D.J.; Hynynen, K. Focused ultrasound-mediated bbb disruption is associated with an increase in activation of AKT: Experimental study in rats. BMC Neurol. 2010, 10, 114. [Google Scholar] [CrossRef]
- Liu, H.-L.; Huang, C.-Y.; Chen, J.-Y.; Wang, H.-Y.J.; Chen, P.-Y.; Wei, K.-C. Pharmacodynamic and Therapeutic Investigation of Focused Ultrasound-Induced Blood-Brain Barrier Opening for Enhanced Temozolomide Delivery in Glioma Treatment. PLoS ONE 2014, 9, e114311. [Google Scholar] [CrossRef] [PubMed]
- Beccaria, K.; Canney, M.; Goldwirt, L.; Fernandez, C.; Piquet, J.; Perier, M.-C.; Lafon, C.; Chapelon, J.-Y.; Carpentier, A. Ultrasound-induced opening of the blood-brain barrier to enhance temozolomide and irinotecan delivery: An experimental study in rabbits. J. Neurosurg. JNS 2016, 124, 1602–1610. [Google Scholar] [CrossRef]
- Chen, H.; Konofagou, E.E. The Size of Blood–Brain Barrier Opening Induced by Focused Ultrasound is Dictated by the Acoustic Pressure. J. Cereb. Blood Flow Metab. 2014, 34, 1197–1204. [Google Scholar] [CrossRef]
- Patel, R.R.; Mehta, M.P. Targeted Therapy for Brain Metastases: Improving the Therapeutic Ratio. Clin. Cancer Res. 2007, 13, 1675–1683. [Google Scholar] [CrossRef]
- Sloan, A.E.; Ahluwalia, M.S.; Valerio-Pascua, J.; Manjila, S.; Torchia, M.G.; Jones, S.E.; Sunshine, J.L.; Phillips, M.; Griswold, M.A.; Clampitt, M.; et al. Results of the NeuroBlate System first-in-humans Phase I clinical trial for recurrent glioblastoma: Clinical article. J. Neurosurg. JNS 2013, 118, 1202–1219. [Google Scholar] [CrossRef]
Cancer Type | Drug | Combination Therapy | Target | Corresponding Effects | Limitation | Ref. |
---|---|---|---|---|---|---|
Metastatic breast cancer | Bevacizumab | Paclitaxel | VEGF-A | Significantly prolonged progression-free survival as compared with paclitaxel alone and increased the objective response rate. | No change in overall survival. | [121] |
NSCLC | Bevaci-zumab | Cisplatin Gemcitabine (CG) | VEGF-A | Combining bevacizumab (7.5 or 15 mg/kg) with CG significantly improved PFS and an objective response rate. | The overall incidence of serious adverse events was higher in the combination group than in the control group. | [122] |
HER2-negative breast cancer | Bevaci-zumab | Adriamycin Cyclophosphamide Paclitaxel (ACP) | VEGF-A | Bevacizumab combined with ACP showed superior anti-tumor activity in TNBC. Bevacizumab induced changes in the vascular structure and levels of circulating biomarkers indicative of vascular normalization in breast cancer. | Tumor regression from the VEGF blockade might be restricted to tumors with a sufficiently high MVD. | [123] |
Glioblastoma | Ziv-aflibercept | γ radiation | VEGFR-2 | The VEGFR-2 blockade can temporarily normalize the tumor vessel structure (pericyte and basement membrane coverage), leading to improved vascular function (tumor oxygenation) and an enhanced response to radiation therapy. | / | [124,125] |
Glioma | Sunitinib | Temozolomide | VEGFR | Sunitinib decreased tumor IFP and increased temozolomide delivery. For mothiazole amine and PVC, it is closely relative to coverage, and its transmission efficiency is inversely proportional to the density of the collagen type IV basement membrane, showing that it can induce the normalization of tumor blood vessels. | / | [126,127] |
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. |
© 2024 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
Zhu, S.; Wang, X.; Jiang, H. Systematic Reversal of Drug Resistance in Cancer. Targets 2024, 2, 250-286. https://doi.org/10.3390/targets2030015
Zhu S, Wang X, Jiang H. Systematic Reversal of Drug Resistance in Cancer. Targets. 2024; 2(3):250-286. https://doi.org/10.3390/targets2030015
Chicago/Turabian StyleZhu, Shujie, Xuemei Wang, and Hui Jiang. 2024. "Systematic Reversal of Drug Resistance in Cancer" Targets 2, no. 3: 250-286. https://doi.org/10.3390/targets2030015
APA StyleZhu, S., Wang, X., & Jiang, H. (2024). Systematic Reversal of Drug Resistance in Cancer. Targets, 2(3), 250-286. https://doi.org/10.3390/targets2030015