Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Debate

Tracing the path of cancer initiation: the AA protein-based model for cancer genesis

Author: Adouda Adjiri

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

Cancer is a defiant disease which cure is still far from being attained besides the colossal efforts and financial means deployed towards that end. The continuing setbacks encountered with today’s arsenal of anti-cancer drugs and cancer therapy modalities; show the need for a radical approach in order to get to the root of the problem. And getting to the root of cancer initiation and development leads us to challenge the present dogmas surrounding the pathogenesis of this disease.

Results

This comprehensive analysis brings to light the following points: (i) Cancer with its plethora of genetic and cellular symptoms could originate from one major event switching a cell from normalcy-to-malignancy; (ii) The switching event is postulated to involve a pathological breakup of a non-mutated protein, called here AA protein, resulting in the acquisition of new cellular functions present only in cancer cells; (iii) Following this event, DNA mutations begin to accumulate as secondary events to ensure perpetuity of cancer. Supporting arguments for this protein-based model come mainly from these observations: (i) The AA protein-based model reconciles together the clonal-and-stem cell theories into one inclusive model; (ii) The breakup of a normal protein could be behind the cancer-linked inflammation symptom; (iii) Cancer hallmarks are but adaptive traits, earned as a result of the switch from normalcy-to-malignancy.

Conclusions

Adaptation of cancer cells to their microenvironment and to different anti-cancer drugs is deemed here as the ultimate cancer hallmark, that needs to be understood and controlled. This adaptive power of cancer cells parallels that of bacteria also known with their resistance to a large range of substances in nature and in the laboratory. Consequently, cancer development could be viewed as a backward walk on the line of Evolution. Finally this unprecedented analysis demystifies cancer and puts the finger on the core problem of malignancy while offering ideas for its control with the ultimate goal of leading to its cure.
Literature
1.
go back to reference Bray F, Jemal A, Grey N, et al. Global cancer transitions according to the human development index (2008-2030): a population-based study. Lancet Oncol. 2012;13:790–801.PubMedCrossRef Bray F, Jemal A, Grey N, et al. Global cancer transitions according to the human development index (2008-2030): a population-based study. Lancet Oncol. 2012;13:790–801.PubMedCrossRef
4.
go back to reference Lord CJ, Ashworth A. Mechanisms of resistance to therapies targeting BRCA-mutant cancers. Nat Med. 2013;19:1381–8.PubMedCrossRef Lord CJ, Ashworth A. Mechanisms of resistance to therapies targeting BRCA-mutant cancers. Nat Med. 2013;19:1381–8.PubMedCrossRef
5.
go back to reference Bouwman P, Jonkers J. Molecular pathways: how can BRCA-mutated tumors become resistant to PARP inhibitors? Clin Cancer Res. 2014;20:540–7.PubMedCrossRef Bouwman P, Jonkers J. Molecular pathways: how can BRCA-mutated tumors become resistant to PARP inhibitors? Clin Cancer Res. 2014;20:540–7.PubMedCrossRef
6.
go back to reference Silk AD, Zasadil LM, Holland AJ, et al. Chromosome mis-segregation rate predicts whether aneuploidy will promote or suppress tumors. Proc Natl Acad Sci U S A. 2013;110(44):E4134–41.PubMedPubMedCentralCrossRef Silk AD, Zasadil LM, Holland AJ, et al. Chromosome mis-segregation rate predicts whether aneuploidy will promote or suppress tumors. Proc Natl Acad Sci U S A. 2013;110(44):E4134–41.PubMedPubMedCentralCrossRef
8.
go back to reference Hahn WC, Weinberg RA. Modeling the molecular circuitry of cancer. Nature Rev Cancer. 2002;2(5):331–41.CrossRef Hahn WC, Weinberg RA. Modeling the molecular circuitry of cancer. Nature Rev Cancer. 2002;2(5):331–41.CrossRef
14.
go back to reference Miller TW, Rexer BN, Garrett JT, et al. Mutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res. 2011;13:224.PubMedPubMedCentralCrossRef Miller TW, Rexer BN, Garrett JT, et al. Mutations in the phosphatidylinositol 3-kinase pathway: role in tumor progression and therapeutic implications in breast cancer. Breast Cancer Res. 2011;13:224.PubMedPubMedCentralCrossRef
17.
go back to reference Wang TJ, Huang MS, Hong CY, et al. Comparisons of tumor suppressor p53, p21, and p16 gene therapy effects on glioblastoma Tumorigenicity in Situ. Biohem Biophys Res Commun. 2001;287 (1:173–80.CrossRef Wang TJ, Huang MS, Hong CY, et al. Comparisons of tumor suppressor p53, p21, and p16 gene therapy effects on glioblastoma Tumorigenicity in Situ. Biohem Biophys Res Commun. 2001;287 (1:173–80.CrossRef
22.
go back to reference Janiec-Jankowska A, Konopka B, Goluda C, et al. TP53 mutations in endometrial cancers: relation to PTEN gene defects. Int J Gynecol Cancer. 2010;20(2):196–202.PubMedCrossRef Janiec-Jankowska A, Konopka B, Goluda C, et al. TP53 mutations in endometrial cancers: relation to PTEN gene defects. Int J Gynecol Cancer. 2010;20(2):196–202.PubMedCrossRef
24.
go back to reference He TC, Sparks AB, Rago C, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281(5382):1509–12.PubMedCrossRef He TC, Sparks AB, Rago C, et al. Identification of c-MYC as a target of the APC pathway. Science. 1998;281(5382):1509–12.PubMedCrossRef
25.
go back to reference van de Wetering M, de Lau W, Clevers H. WNT signaling and lymphocyte development. Cell. 2002;109:S13–9.PubMedCrossRef van de Wetering M, de Lau W, Clevers H. WNT signaling and lymphocyte development. Cell. 2002;109:S13–9.PubMedCrossRef
27.
go back to reference Gauthier ML, Berman HK, Miller C, et al. Abrogated response to cellular stress identifies DCIS associated with subsequent tumor events and defines basal-like breast tumors. Cancer Cell. 2007;12(5):479–91.PubMedPubMedCentralCrossRef Gauthier ML, Berman HK, Miller C, et al. Abrogated response to cellular stress identifies DCIS associated with subsequent tumor events and defines basal-like breast tumors. Cancer Cell. 2007;12(5):479–91.PubMedPubMedCentralCrossRef
28.
go back to reference Lewis CM, Cler LR, Bu DW, et al. Promoter Hypermethylation in Benign Breast Epithelium in Relation to Predicted Breast Cancer Risk. Clin Cancer Res. 2005;11:166–72. Lewis CM, Cler LR, Bu DW, et al. Promoter Hypermethylation in Benign Breast Epithelium in Relation to Predicted Breast Cancer Risk. Clin Cancer Res. 2005;11:166–72.
29.
go back to reference Yan PS, Venkataramu C, Ibrahim A, et al. Mapping geographic zones of cancer risk with epigenetic biomarkers in normal breast tissue. Clin Cancer Res. 2006;12(22):6626–36.PubMedCrossRef Yan PS, Venkataramu C, Ibrahim A, et al. Mapping geographic zones of cancer risk with epigenetic biomarkers in normal breast tissue. Clin Cancer Res. 2006;12(22):6626–36.PubMedCrossRef
31.
go back to reference Wood LD, Parsons DW, Jones S, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318(5853):8–9.CrossRef Wood LD, Parsons DW, Jones S, et al. The genomic landscapes of human breast and colorectal cancers. Science. 2007;318(5853):8–9.CrossRef
33.
go back to reference Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8:729–40.PubMedCrossRef Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8:729–40.PubMedCrossRef
34.
go back to reference Gosselin K, Martien S, Pourtier A, et al. Senescence-associated oxidative DNA damage promotes the generation of neoplastic cells. Cancer Res. 2009;69:7917–25.PubMedCrossRef Gosselin K, Martien S, Pourtier A, et al. Senescence-associated oxidative DNA damage promotes the generation of neoplastic cells. Cancer Res. 2009;69:7917–25.PubMedCrossRef
35.
38.
go back to reference Chung HY, Cesari M, Anton S, et al. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev. 2009;8:18–30.PubMedCrossRef Chung HY, Cesari M, Anton S, et al. Molecular inflammation: underpinnings of aging and age-related diseases. Ageing Res Rev. 2009;8:18–30.PubMedCrossRef
41.
go back to reference Muller D, Breathnach R, Engelmann A, et al. Expression of collagenase-related metalloproteinase genes in human lung or head and neck tumours. Int J Cancer. 1991;48:550–6.PubMedCrossRef Muller D, Breathnach R, Engelmann A, et al. Expression of collagenase-related metalloproteinase genes in human lung or head and neck tumours. Int J Cancer. 1991;48:550–6.PubMedCrossRef
42.
go back to reference Wolf C, Rovyer N. Stromelysin-3 belongs to a subgroup of proteinases expressed in breast carcinoma fibroblastic cells and possibly implicated in tumour progression. Proc Natl Acad Sci U S A. 1993;90:1843–7.PubMedPubMedCentralCrossRef Wolf C, Rovyer N. Stromelysin-3 belongs to a subgroup of proteinases expressed in breast carcinoma fibroblastic cells and possibly implicated in tumour progression. Proc Natl Acad Sci U S A. 1993;90:1843–7.PubMedPubMedCentralCrossRef
43.
go back to reference Newell KJ, Witty JP, Rodgers WH, et al. Expression and localisation of matrix-degrading metalloproteinases during colorectal tumourigenesis. Mol Carcinog. 1994;10:199–206.PubMedCrossRef Newell KJ, Witty JP, Rodgers WH, et al. Expression and localisation of matrix-degrading metalloproteinases during colorectal tumourigenesis. Mol Carcinog. 1994;10:199–206.PubMedCrossRef
44.
go back to reference Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2:161–74.PubMedCrossRef Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression. Nat Rev Cancer. 2002;2:161–74.PubMedCrossRef
45.
go back to reference Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–9.PubMedCrossRef Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–9.PubMedCrossRef
48.
go back to reference Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–8.PubMedCrossRef Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer. 2004;4:71–8.PubMedCrossRef
49.
go back to reference Roberts CK, Sindhu KK. Oxidative stress and metabolic syndrome. Life Sci. 2009;84(21–22):705–12.PubMedCrossRef Roberts CK, Sindhu KK. Oxidative stress and metabolic syndrome. Life Sci. 2009;84(21–22):705–12.PubMedCrossRef
50.
go back to reference Sedelnikova OA, Horikawa I, Zimonjic DB, et al. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double strand breaks. Nat Cell Biol. 2004;6(2):168–70.PubMedCrossRef Sedelnikova OA, Horikawa I, Zimonjic DB, et al. Senescing human cells and ageing mice accumulate DNA lesions with unrepairable double strand breaks. Nat Cell Biol. 2004;6(2):168–70.PubMedCrossRef
51.
go back to reference Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection? Nucleic Acids Res. 2007;35(22):7505–13.PubMedPubMedCentralCrossRef Passos JF, Saretzki G, von Zglinicki T. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection? Nucleic Acids Res. 2007;35(22):7505–13.PubMedPubMedCentralCrossRef
52.
go back to reference Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004;6:463–77.PubMedCrossRef Levine B, Klionsky DJ. Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 2004;6:463–77.PubMedCrossRef
56.
go back to reference Degenhardt K, Chen G, Lindsten T, et al. BAX and BAK mediate p53-independent suppression of tumorigenesis. Cancer Cell. 2002;2:193–203.PubMedCrossRef Degenhardt K, Chen G, Lindsten T, et al. BAX and BAK mediate p53-independent suppression of tumorigenesis. Cancer Cell. 2002;2:193–203.PubMedCrossRef
57.
60.
go back to reference Rodriguez OC, Choudhury S, Kolukula V, et al. Dietary down-regulation of mutant p53 levels via glucose restriction: mechanisms and implications for tumor therapy. Cell Cycle. 2012;11:4436–46.PubMedPubMedCentralCrossRef Rodriguez OC, Choudhury S, Kolukula V, et al. Dietary down-regulation of mutant p53 levels via glucose restriction: mechanisms and implications for tumor therapy. Cell Cycle. 2012;11:4436–46.PubMedPubMedCentralCrossRef
61.
go back to reference Choudhury S, Kolukula VK, Preet A, et al. Dissecting the pathways that destabilize mutant p53: the proteasome or autophagy? Cell Cycle. 2013;12:1022–9.PubMedPubMedCentralCrossRef Choudhury S, Kolukula VK, Preet A, et al. Dissecting the pathways that destabilize mutant p53: the proteasome or autophagy? Cell Cycle. 2013;12:1022–9.PubMedPubMedCentralCrossRef
63.
go back to reference Lazova R, Camp RL, Klump V, et al. Punctate LC3B expression is a common feature of solid tumors and associated with proliferation, metastasis, and poor outcome. Clin Cancer Res. 2012;18:370–9.PubMedCrossRef Lazova R, Camp RL, Klump V, et al. Punctate LC3B expression is a common feature of solid tumors and associated with proliferation, metastasis, and poor outcome. Clin Cancer Res. 2012;18:370–9.PubMedCrossRef
71.
go back to reference Reya T, Morrison SJ, Clarke MF, et al. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMedCrossRef Reya T, Morrison SJ, Clarke MF, et al. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11.PubMedCrossRef
72.
77.
go back to reference Shackleton M, Quintana E, Fearon ER, et al. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell. 2009;138:822–9.PubMedCrossRef Shackleton M, Quintana E, Fearon ER, et al. Heterogeneity in cancer: cancer stem cells versus clonal evolution. Cell. 2009;138:822–9.PubMedCrossRef
78.
go back to reference Quintana E, Shackleton M, Foster HR, et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010;18:510–23.PubMedPubMedCentralCrossRef Quintana E, Shackleton M, Foster HR, et al. Phenotypic heterogeneity among tumorigenic melanoma cells from patients that is reversible and not hierarchically organized. Cancer Cell. 2010;18:510–23.PubMedPubMedCentralCrossRef
79.
go back to reference Campbell LL, Polyak K. Breast tumor heterogeneity: cancer stem cells or clonal evolution? Cell Cycle. 2007;6:2332–8.PubMedCrossRef Campbell LL, Polyak K. Breast tumor heterogeneity: cancer stem cells or clonal evolution? Cell Cycle. 2007;6:2332–8.PubMedCrossRef
80.
go back to reference Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–68.PubMedCrossRef Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8:755–68.PubMedCrossRef
81.
go back to reference Vermeulen L, Sprick MR, Kemper K, et al. Cancer stem cells – old concepts, new insights. Cell Death Differ. 2008;15:947–58.PubMedCrossRef Vermeulen L, Sprick MR, Kemper K, et al. Cancer stem cells – old concepts, new insights. Cell Death Differ. 2008;15:947–58.PubMedCrossRef
83.
go back to reference Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature. 2005;438:820–7.PubMedPubMedCentralCrossRef Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature. 2005;438:820–7.PubMedPubMedCentralCrossRef
84.
go back to reference Talmadge JE, Wolman SR, Fidler IJ. Evidence for the clonal origin of spontaneous metastases. Science. 1982;217:361–3.PubMedCrossRef Talmadge JE, Wolman SR, Fidler IJ. Evidence for the clonal origin of spontaneous metastases. Science. 1982;217:361–3.PubMedCrossRef
86.
go back to reference Mueller MM, Fusenig NE. Friends or foes—bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004;4:839–49.PubMedCrossRef Mueller MM, Fusenig NE. Friends or foes—bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004;4:839–49.PubMedCrossRef
89.
go back to reference Wyckoff B, Wang Y, Lin EY, et al. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67:2649–56.PubMedCrossRef Wyckoff B, Wang Y, Lin EY, et al. Direct visualization of macrophage-assisted tumor cell intravasation in mammary tumors. Cancer Res. 2007;67:2649–56.PubMedCrossRef
90.
go back to reference Paget S. The distribution of secondary growths in cancer of the breast. Lancet. 1889;1:571–3.CrossRef Paget S. The distribution of secondary growths in cancer of the breast. Lancet. 1889;1:571–3.CrossRef
91.
go back to reference Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3:453–8.PubMedCrossRef Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3:453–8.PubMedCrossRef
92.
go back to reference Sullivan R, Graham CH. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007;26:319–31.PubMedCrossRef Sullivan R, Graham CH. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007;26:319–31.PubMedCrossRef
93.
go back to reference Kang TW, Yevsa T, Woller N, et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature. 2011;479:547–51.PubMedCrossRef Kang TW, Yevsa T, Woller N, et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature. 2011;479:547–51.PubMedCrossRef
95.
go back to reference Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–54.PubMedCrossRef Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2:442–54.PubMedCrossRef
96.
go back to reference Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7:131–42.PubMedCrossRef Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7:131–42.PubMedCrossRef
97.
go back to reference Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3:362–74.PubMedCrossRef Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3:362–74.PubMedCrossRef
98.
go back to reference Wolf K, Mazo I, Leung H, et al. Compensation mechanism in tumor cell migration: mesenchymal–amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 2003;160:267–77.PubMedPubMedCentralCrossRef Wolf K, Mazo I, Leung H, et al. Compensation mechanism in tumor cell migration: mesenchymal–amoeboid transition after blocking of pericellular proteolysis. J Cell Biol. 2003;160:267–77.PubMedPubMedCentralCrossRef
101.
go back to reference Baer CF, Miyamoto MM, Denver DR. Mutation rate variation in multicellular eukaryotes: causes and consequences. Nature Rev Genet. 2007;8:619–31.PubMedCrossRef Baer CF, Miyamoto MM, Denver DR. Mutation rate variation in multicellular eukaryotes: causes and consequences. Nature Rev Genet. 2007;8:619–31.PubMedCrossRef
104.
go back to reference Bates RC, Edwards NS, Yates JD. Spheroids and cell survival. Crit Rev Oncol Hematol. 2000;36:61–74.PubMedCrossRef Bates RC, Edwards NS, Yates JD. Spheroids and cell survival. Crit Rev Oncol Hematol. 2000;36:61–74.PubMedCrossRef
105.
go back to reference He Z, Kannan N, Nemirovsky O, et al. BRCA1 controls the cell division axis and governs ploidy and phenotype in human mammary cells. Oncotarget. 2017;8(20):32461–75.PubMedPubMedCentral He Z, Kannan N, Nemirovsky O, et al. BRCA1 controls the cell division axis and governs ploidy and phenotype in human mammary cells. Oncotarget. 2017;8(20):32461–75.PubMedPubMedCentral
106.
go back to reference Morroni M, Giordano A, Zingaretti MC, et al. Reversible trans-differentiation of secretory epithelial cells into adipocytes in the mammary gland. Proc Natl Acad Sci U S A. 2004;101:16801–6.PubMedPubMedCentralCrossRef Morroni M, Giordano A, Zingaretti MC, et al. Reversible trans-differentiation of secretory epithelial cells into adipocytes in the mammary gland. Proc Natl Acad Sci U S A. 2004;101:16801–6.PubMedPubMedCentralCrossRef
108.
go back to reference Ben-Porath I, Weinberg RA. The signals and pathways activating cellular senescence. Int J Biochem Cell Biol. 2005;37(5):961–76.PubMedCrossRef Ben-Porath I, Weinberg RA. The signals and pathways activating cellular senescence. Int J Biochem Cell Biol. 2005;37(5):961–76.PubMedCrossRef
109.
go back to reference Dolcetti R, de Rossi A. Telomere/telomerase interplay in virus-driven and virus-independent lymphomagenesis: pathogenic and clinical implications. Med Res Rev. 2012;32:233–53.PubMedCrossRef Dolcetti R, de Rossi A. Telomere/telomerase interplay in virus-driven and virus-independent lymphomagenesis: pathogenic and clinical implications. Med Res Rev. 2012;32:233–53.PubMedCrossRef
Metadata
Title
Tracing the path of cancer initiation: the AA protein-based model for cancer genesis
Author
Adouda Adjiri
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4739-1

Other articles of this Issue 1/2018

BMC Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine