Neural induction drives body axis formation during embryogenesis, but a neural induction-like process drives tumorigenesis in postnatal animals

Characterization of cancer cells and neural stem cells indicates that tumorigenicity and pluripotency are coupled cell properties determined by neural stemness, and tumorigenesis represents a process of progressive loss of original cell identity and gain of neural stemness. This reminds of a most fundamental process required for the development of the nervous system and body axis during embryogenesis, i.e., embryonic neural induction. Neural induction is that, in response to extracellular signals that are secreted by the Spemann-Mangold organizer in amphibians or the node in mammals and inhibit epidermal fate in ectoderm, the ectodermal cells lose their epidermal fate and assume the neural default fate and consequently, turn into neuroectodermal cells. They further differentiate into the nervous system and also some non-neural cells via interaction with adjacent tissues. Failure in neural induction leads to failure of embryogenesis, and ectopic neural induction due to ectopic organizer or node activity or activation of embryonic neural genes causes a formation of secondary body axis or a conjoined twin. During tumorigenesis, cells progressively lose their original cell identity and gain of neural stemness, and consequently, gain of tumorigenicity and pluripotency, due to various intra-/extracellular insults in cells of a postnatal animal. Tumorigenic cells can be induced to differentiation into normal cells and integrate into normal embryonic development within an embryo. However, they form tumors and cannot integrate into animal tissues/organs in a postnatal animal because of lack of embryonic inducing signals. Combination of studies of developmental and cancer biology indicates that neural induction drives embryogenesis in gastrulating embryos but a similar process drives tumorigenesis in a postnatal animal. Tumorigenicity is by nature the manifestation of aberrant occurrence of pluripotent state in a postnatal animal. Pluripotency and tumorigenicity are both but different manifestations of neural stemness in pre- and postnatal stages of animal life, respectively. Based on these findings, I discuss about some confusion in cancer research, propose to distinguish the causality and associations and discriminate causal and supporting factors involved in tumorigenesis, and suggest revisiting the focus of cancer research.

[1]  T. Kuner,et al.  Glioblastoma hijacks neuronal mechanisms for brain invasion , 2022, Cell.

[2]  P. Saw,et al.  Targeting CAFs to overcome anticancer therapeutic resistance. , 2022, Trends in cancer.

[3]  N. Normanno,et al.  In PD-1+ human colon cancer cells NIVOLUMAB promotes survival and could protect tumor cells from conventional therapies , 2022, Journal for ImmunoTherapy of Cancer.

[4]  J. Creemers,et al.  The proprotein convertase furin in cancer: more than an oncogene , 2022, Oncogene.

[5]  M. Zhang,et al.  Neural stemness unifies cell tumorigenicity and pluripotent differentiation potential , 2021, bioRxiv.

[6]  N. Callewaert,et al.  T Cell Engaging Immunotherapies, Highlighting Chimeric Antigen Receptor (CAR) T Cell Therapy , 2021, Cancers.

[7]  A. Yoshimi,et al.  Aberrant RNA splicing and therapeutic opportunities in cancers , 2021, Cancer science.

[8]  Xuehao Wang,et al.  Targeting Immune Cells in the Tumor Microenvironment of HCC: New Opportunities and Challenges , 2021, Frontiers in Cell and Developmental Biology.

[9]  A. Shilatifard,et al.  Dietary palmitic acid promotes a prometastatic memory via Schwann cells , 2021, Nature.

[10]  Ying Cao Neural is Fundamental: Neural Stemness as the Ground State of Cell Tumorigenicity and Differentiation Potential , 2021, Stem Cell Reviews and Reports.

[11]  Ying Cao,et al.  Coordinated regulation of the ribosome and proteasome by PRMT1 in the maintenance of neural stemness in cancer cells and neural stem cells , 2021, The Journal of biological chemistry.

[12]  J. de Magalhães Every gene can (and possibly will) be associated with cancer. , 2021, Trends in genetics : TIG.

[13]  Ying Cao,et al.  Suppression of Cell Tumorigenicity by Non-neural Pro-differentiation Factors via Inhibition of Neural Property in Tumorigenic Cells , 2021, Frontiers in Cell and Developmental Biology.

[14]  C. Galassi,et al.  The Immune Privilege of Cancer Stem Cells: A Key to Understanding Tumor Immune Escape and Therapy Failure , 2021, Cells.

[15]  Z. Tian,et al.  Rectal adenocarcinoma with multifocal osteoid differentiation. , 2021, Asian Journal of Surgery.

[16]  S. Sadeghi,et al.  Battling Chemoresistance in Cancer: Root Causes and Strategies to Uproot Them , 2021, International journal of molecular sciences.

[17]  T. Brabletz,et al.  Dynamic EMT: a multi‐tool for tumor progression , 2021, The EMBO journal.

[18]  S. Wiemann,et al.  Cancer-Associated Fibroblasts: Implications for Cancer Therapy , 2021, Cancers.

[19]  T. Lee,et al.  Cancer Stem Cells: Emerging Key Players in Immune Evasion of Cancers , 2021, Frontiers in Cell and Developmental Biology.

[20]  T. Sellers,et al.  Morphologic and molecular correlates of EZH2 as a predictor of platinum resistance in high-grade ovarian serous carcinoma , 2021, BMC cancer.

[21]  Y. Buganim,et al.  Targeting neurons in the tumor microenvironment with bupivacaine nanoparticles reduces breast cancer progression and metastases , 2021, bioRxiv.

[22]  K. Bussey,et al.  Cancer progression as a sequence of atavistic reversions , 2021, BioEssays : news and reviews in molecular, cellular and developmental biology.

[23]  Yvonne Y. Chen,et al.  Navigating CAR-T cells through the solid-tumour microenvironment , 2021, Nature Reviews Drug Discovery.

[24]  Jacob D. Jaffe,et al.  Epigenetic silencing by SETDB1 suppresses tumour intrinsic immunogenicity , 2021, Nature.

[25]  R. Weinberg,et al.  Linking EMT programmes to normal and neoplastic epithelial stem cells , 2021, Nature Reviews Cancer.

[26]  Rasheed Omobolaji Alabi,et al.  Clinical significance of tumor-stroma ratio in head and neck cancer: a systematic review and meta-analysis , 2021, BMC cancer.

[27]  Rosalie M Sterner,et al.  CAR-T cell therapy: current limitations and potential strategies , 2021, Blood Cancer Journal.

[28]  M. Ferrer,et al.  Isolating and targeting the real-time plasticity and malignant properties of epithelial-mesenchymal transition in cancer , 2021, Oncogene.

[29]  Robin L. Jones,et al.  Extraskeletal osteosarcomas: current update. , 2021, Future oncology.

[30]  S. Loi,et al.  Intratumoral heterogeneity in cancer progression and response to immunotherapy , 2021, Nature Medicine.

[31]  H. Pijl,et al.  Tumor‐stroma ratio is associated with Miller‐Payne score and pathological response to neoadjuvant chemotherapy in HER2‐negative early breast cancer , 2020, International journal of cancer.

[32]  Ying Cao,et al.  Neural stemness contributes to cell tumorigenicity , 2020, Cell & Bioscience.

[33]  R. Sambasivan,et al.  Neuromesodermal Progenitors: A Basis for Robust Axial Patterning in Development and Evolution , 2021, Frontiers in Cell and Developmental Biology.

[34]  G. Calin,et al.  Cancer-Associated Neurogenesis and Nerve-Cancer Cross-talk , 2020, Cancer Research.

[35]  É. Vivier,et al.  Tumor-Infiltrating Natural Killer Cells. , 2020, Cancer discovery.

[36]  K. Lehti,et al.  Crosstalk Between Cancer Associated Fibroblasts and Cancer Cells in Scirrhous Type Gastric Cancer , 2020, Frontiers in Oncology.

[37]  Katherine Cosmopoulos,et al.  Overcoming Immune Checkpoint Blockade Resistance via EZH2 Inhibition. , 2020, Trends in immunology.

[38]  F. Marcucci,et al.  The tumor-promoting effects of the adaptive immune system: a cause of hyperprogressive disease in cancer? , 2020, Cellular and Molecular Life Sciences.

[39]  R. Drapkin,et al.  Tumor Innervation: Cancer Has Some Nerve. , 2020, Trends in cancer.

[40]  A. Rustgi,et al.  EMT, MET, Plasticity, and Tumor Metastasis. , 2020, Trends in cell biology.

[41]  E. Calvo,et al.  Clinical Challenges of Immune Checkpoint Inhibitors. , 2020, Cancer cell.

[42]  B. Stanger,et al.  How Tumor Cell Dedifferentiation Drives Immune Evasion and Resistance to Immunotherapy , 2020, Cancer Research.

[43]  Rodolfo Daniel Cervantes-Villagrana,et al.  Tumor-induced neurogenesis and immune evasion as targets of innovative anti-cancer therapies , 2020, Signal Transduction and Targeted Therapy.

[44]  M. K. Siu,et al.  Infiltration of T cells promotes the metastasis of ovarian cancer cells via the modulation of metastasis-related genes and PD-L1 expression , 2020, Cancer Immunology, Immunotherapy.

[45]  D. Tuveson,et al.  DIVERSITY AND BIOLOGY OF CANCER-ASSOCIATED FIBROBLASTS. , 2020, Physiological reviews.

[46]  I. Aifantis,et al.  RNA Splicing and Cancer. , 2020, Trends in cancer.

[47]  Joshua M. Weiss,et al.  Developmental chromatin programs determine oncogenic competence in melanoma , 2020, bioRxiv.

[48]  Xi C. He,et al.  Overcoming Wnt–β-catenin dependent anticancer therapy resistance in leukaemia stem cells , 2020, Nature Cell Biology.

[49]  Raymond B. Runyan,et al.  Guidelines and definitions for research on epithelial–mesenchymal transition , 2020, Nature Reviews Molecular Cell Biology.

[50]  M. Hellmann,et al.  Acquired Resistance to Immune Checkpoint Inhibitors. , 2020, Cancer cell.

[51]  G. Gao,et al.  Tumor cell-intrinsic PD-1 receptor is a tumor suppressor and mediates resistance to PD-1 blockade therapy , 2020, Proceedings of the National Academy of Sciences.

[52]  D. Pe’er,et al.  Lineage plasticity in cancer: a shared pathway of therapeutic resistance , 2020, Nature Reviews Clinical Oncology.

[53]  K. Zilles,et al.  Neurodevelopment , 2020, New Oxford Textbook of Psychiatry.

[54]  Masahide Takahashi,et al.  Cancer‐associated fibroblasts that restrain cancer progression: Hypotheses and perspectives , 2020, Cancer science.

[55]  K. D. de Visser,et al.  Immune crosstalk in cancer progression and metastatic spread: a complex conversation , 2020, Nature Reviews Immunology.

[56]  T. Cai,et al.  A Switch in Tissue Stem Cell Identity Causes Neuroendocrine Tumors in Drosophila Gut. , 2020, Cell reports.

[57]  Jing Wang,et al.  Loss of p53 drives neuron reprogramming in head and neck cancer , 2020, Nature.

[58]  H. Putter,et al.  The prognostic value of the tumor–stroma ratio is most discriminative in patients with grade III or triple‐negative breast cancer , 2020, International journal of cancer.

[59]  I. Kozeretska,et al.  BRCA1 and EZH2 cooperate in regulation of prostate cancer stem cell phenotype , 2019, International journal of cancer.

[60]  A. Dimberg,et al.  Tumor angiogenesis: causes, consequences, challenges and opportunities , 2019, Cellular and Molecular Life Sciences.

[61]  T. Tajiri,et al.  Derivation of neural stem cells from human teratomas. , 2019, Stem cell research.

[62]  A. Ribas,et al.  Tumour-intrinsic resistance to immune checkpoint blockade , 2019, Nature Reviews Immunology.

[63]  Xiaojing Ma,et al.  Targeting EZH2 Enhances Antigen Presentation, Antitumor Immunity, and Circumvents Anti–PD-1 Resistance in Head and Neck Cancer , 2019, Clinical Cancer Research.

[64]  Hsi-Wen Yeh,et al.  A New Switch for TGFβ in Cancer. , 2019, Cancer research.

[65]  James R. Anderson,et al.  Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. , 2019, The Lancet. Oncology.

[66]  L. Shevde,et al.  The Tumor Microenvironment Innately Modulates Cancer Progression. , 2019, Cancer research.

[67]  E. D. De Robertis,et al.  Transcriptome analysis of regeneration during Xenopus laevis experimental twinning. , 2019, The International journal of developmental biology.

[68]  M. Merad,et al.  β-catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. , 2019, Cancer discovery.

[69]  E. Fuchs,et al.  Adaptive Immune Resistance Emerges from Tumor-Initiating Stem Cells , 2019, Cell.

[70]  Ying Cao,et al.  EZH2 Regulates Protein Stability via Recruiting USP7 to Mediate Neuronal Gene Expression in Cancer Cells , 2019, Front. Genet..

[71]  Y. Allory,et al.  Progenitors from the central nervous system drive neurogenesis in cancer , 2019, Nature.

[72]  H. Lamb,et al.  Primary Osteosarcoma of the Breast. , 2019, Radiographics : a review publication of the Radiological Society of North America, Inc.

[73]  Hiromasa Morikawa,et al.  PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer , 2019, Proceedings of the National Academy of Sciences.

[74]  B. Stanger,et al.  Cellular Plasticity in Cancer. , 2019, Cancer discovery.

[75]  Michael Bordonaro,et al.  Quantum biology and human carcinogenesis , 2019, Biosyst..

[76]  R. Tollenaar,et al.  Increased expression of cancer-associated fibroblast markers at the invasive front and its association with tumor-stroma ratio in colorectal cancer , 2019, BMC cancer.

[77]  Masahide Takahashi,et al.  Cancer-associated fibroblasts in gastrointestinal cancer , 2019, Nature Reviews Gastroenterology & Hepatology.

[78]  G. Christofori,et al.  Gain Fat-Lose Metastasis: Converting Invasive Breast Cancer Cells into Adipocytes Inhibits Cancer Metastasis. , 2019, Cancer cell.

[79]  Morris Laster,et al.  When cancer meets quantum mechanics. , 2019, Theoretical biology forum.

[80]  Georgios Giamas,et al.  Astrocytes, the rising stars of the glioblastoma microenvironment , 2018, Glia.

[81]  J. Doench,et al.  Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade , 2018, Nature.

[82]  M. Smyth,et al.  Cancer immunoediting and resistance to T cell-based immunotherapy , 2018, Nature Reviews Clinical Oncology.

[83]  P. Muñoz,et al.  Cancer cell plasticity: Impact on tumor progression and therapy response. , 2018, Seminars in cancer biology.

[84]  A. Monsoro-Burq,et al.  The neural border: Induction, specification and maturation of the territory that generates neural crest cells. , 2018, Developmental biology.

[85]  A. Monsoro-Burq,et al.  AKT signaling displays multifaceted functions in neural crest development. , 2018, Developmental biology.

[86]  J. Soria,et al.  Hyperprogressive disease: recognizing a novel pattern to improve patient management , 2018, Nature Reviews Clinical Oncology.

[87]  M. Donia,et al.  Acquired resistance to cancer immunotherapy , 2018, Seminars in Immunopathology.

[88]  U. Chatterji,et al.  Stemness and chemoresistance are imparted to the OC cells through TGFβ1 driven EMT , 2018, Journal of cellular biochemistry.

[89]  B. Sáinz,et al.  Current perspectives on the crosstalk between lung cancer stem cells and cancer-associated fibroblasts. , 2018, Critical reviews in oncology/hematology.

[90]  Shelly Maman,et al.  A history of exploring cancer in context , 2018, Nature Reviews Cancer.

[91]  A. Martinez Arias,et al.  On the nature and function of organizers , 2018, Development.

[92]  J. Wargo,et al.  The RNA-binding Protein MEX3B Mediates Resistance to Cancer Immunotherapy by Downregulating HLA-A Expression , 2018, Clinical Cancer Research.

[93]  V. Apostolopoulos,et al.  Crosstalk between cancer and the neuro-immune system , 2018, Journal of Neuroimmunology.

[94]  N. Bhardwaj,et al.  Re-Emergence of Dendritic Cell Vaccines for Cancer Treatment. , 2018, Trends in cancer.

[95]  A. Dicker,et al.  Blockade of Tumor-Expressed PD-1 promotes lung cancer growth , 2017, Oncoimmunology.

[96]  H. Thé Differentiation therapy revisited , 2018, Nature Reviews Cancer.

[97]  C. Wright,et al.  Prevention and reversion of pancreatic tumorigenesis through a differentiation-based mechanism , 2017, bioRxiv.

[98]  J. Wilmott,et al.  Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma , 2017, Clinical Cancer Research.

[99]  Ying Cao Tumorigenesis as a process of gradual loss of original cell identity and gain of properties of neural precursor/progenitor cells , 2017, Cell & Bioscience.

[100]  H. Okano,et al.  Low immunogenicity of mouse induced pluripotent stem cell-derived neural stem/progenitor cells , 2017, Scientific Reports.

[101]  C. Lindskog,et al.  A pathology atlas of the human cancer transcriptome , 2017, Science.

[102]  M. Zhang,et al.  Similarity in gene-regulatory networks suggests that cancer cells share characteristics of embryonic neural cells , 2017, The Journal of Biological Chemistry.

[103]  F. Baralle,et al.  Alternative splicing as a regulator of development and tissue identity , 2017, Nature Reviews Molecular Cell Biology.

[104]  Ruth Nussinov,et al.  A New View of Pathway-Driven Drug Resistance in Tumor Proliferation. , 2017, Trends in pharmacological sciences.

[105]  G. Kollias,et al.  Mesenchymal Cells in Colon Cancer. , 2017, Gastroenterology.

[106]  H. Hondermarck,et al.  Nerve Dependence: From Regeneration to Cancer. , 2017, Cancer cell.

[107]  H. Okano,et al.  Evaluation of the immunogenicity of human iPS cell-derived neural stem/progenitor cells in vitro. , 2017, Stem cell research.

[108]  J. Wargo,et al.  Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy , 2017, Cell.

[109]  H. Tomita,et al.  Nerve Growth Factor Promotes Gastric Tumorigenesis through Aberrant Cholinergic Signaling. , 2017, Cancer cell.

[110]  Yanna Shang,et al.  Neurons generated from carcinoma stem cells support cancer progression , 2017, Signal Transduction and Targeted Therapy.

[111]  Araxi O. Urrutia,et al.  Alternative splicing and the evolution of phenotypic novelty , 2016, Philosophical Transactions of the Royal Society B: Biological Sciences.

[112]  C. Welter,et al.  Origin of Cancer: An Information, Energy, and Matter Disease , 2016, Front. Cell Dev. Biol..

[113]  Austin E. Gillen,et al.  Fibroblast Subtypes Regulate Responsiveness of Luminal Breast Cancer to Estrogen , 2016, Clinical Cancer Research.

[114]  J. Hartman,et al.  Pericyte–fibroblast transition promotes tumor growth and metastasis , 2016, Proceedings of the National Academy of Sciences.

[115]  Omar Abdel-Wahab,et al.  Therapeutic targeting of splicing in cancer , 2016, Nature Medicine.

[116]  A. Dutt,et al.  MYOD1 (L122R) mutations are associated with spindle cell and sclerosing rhabdomyosarcomas with aggressive clinical outcomes , 2016, Modern Pathology.

[117]  R. Weichselbaum,et al.  Tumor-associated fibroblasts predominantly come from local and not circulating precursors , 2016, Proceedings of the National Academy of Sciences.

[118]  X. de la Cruz,et al.  EZH2 regulates neuroepithelium structure and neuroblast proliferation by repressing p21 , 2016, Open Biology.

[119]  Aleksandr B. Sahakyan,et al.  Long genes and genes with multiple splice variants are enriched in pathways linked to cancer and other multigenic diseases , 2016, BMC Genomics.

[120]  Craig S. Wong,et al.  Genetic loci associated with renal function measures and chronic kidney disease in children: the Pediatric Investigation for Genetic Factors Linked with Renal Progression Consortium. , 2015, Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association.

[121]  Claire Anderson,et al.  Organizers in Development. , 2016, Current topics in developmental biology.

[122]  D. Henrique,et al.  Neuromesodermal progenitors and the making of the spinal cord , 2015, Development.

[123]  Xiongfong Chen,et al.  The Proto-oncogene Transcription Factor Ets1 Regulates Neural Crest Development through Histone Deacetylase 1 to Mediate Output of Bone Morphogenetic Protein Signaling* , 2015, The Journal of Biological Chemistry.

[124]  AnjaliL . Rao,et al.  Shared regulatory programs suggest retention of blastula-stage potential in neural crest cells , 2015, Science.

[125]  B. Thisse,et al.  Formation of the vertebrate embryo: Moving beyond the Spemann organizer. , 2015, Seminars in cell & developmental biology.

[126]  T. Gajewski,et al.  Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity , 2015, Nature.

[127]  H. Hondermarck,et al.  Nerve-Cancer Cell Cross-talk: A Novel Promoter of Tumor Progression. , 2015, Cancer research.

[128]  N. McGranahan,et al.  Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. , 2015, Cancer cell.

[129]  R. Paduch Theories of cancer origin , 2015, European journal of cancer prevention : the official journal of the European Cancer Prevention Organisation.

[130]  C. Théry,et al.  Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. , 2014, Annual review of cell and developmental biology.

[131]  N. Socci,et al.  A recurrent neomorphic mutation in MYOD1 defines a clinically aggressive subset of embryonal rhabdomyosarcoma associated with PI3K-AKT pathway mutations , 2014, Nature Genetics.

[132]  Tony D. Southall,et al.  Dedifferentiation of Neurons Precedes Tumor Formation in lola Mutants , 2014, Developmental cell.

[133]  R. Weinberg Coming Full Circle—From Endless Complexity to Simplicity and Back Again , 2014, Cell.

[134]  G. Blin,et al.  Bone morphogenic protein signalling suppresses differentiation of pluripotent cells by maintaining expression of E-Cadherin , 2013, eLife.

[135]  C. Blanpain,et al.  Unravelling cancer stem cell potential , 2013, Nature Reviews Cancer.

[136]  N. McGranahan,et al.  The causes and consequences of genetic heterogeneity in cancer evolution , 2013, Nature.

[137]  Corbin E. Meacham,et al.  Tumour heterogeneity and cancer cell plasticity , 2013, Nature.

[138]  S. Freedland,et al.  Autonomic Nerve Development Contributes to Prostate Cancer Progression , 2013, Science.

[139]  D. Nam,et al.  Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. , 2013, Cancer cell.

[140]  R. McLendon,et al.  Glioblastoma Stem Cells Generate Vascular Pericytes to Support Vessel Function and Tumor Growth , 2013, Cell.

[141]  D. Munn,et al.  Indoleamine 2,3 dioxygenase and metabolic control of immune responses. , 2013, Trends in immunology.

[142]  B F Warren,et al.  The proportion of tumor-stroma as a strong prognosticator for stage II and III colon cancer patients: validation in the VICTOR trial. , 2013, Annals of oncology : official journal of the European Society for Medical Oncology.

[143]  N. Harrison,et al.  Rediscovering pluripotency: from teratocarcinomas to embryonic stem cells. Cardiff, 10-12 October 2011. , 2012, The International journal of developmental biology.

[144]  K. Polyak,et al.  Intra-tumour heterogeneity: a looking glass for cancer? , 2012, Nature Reviews Cancer.

[145]  H. Ulrich,et al.  Neural differentiation of P19 carcinoma cells and primary neurospheres: cell morphology, proliferation, viability, and functionality. , 2012, Current protocols in stem cell biology.

[146]  T. Misteli,et al.  In vitro generation of human cells with cancer stem cell properties , 2011, Nature Cell Biology.

[147]  A. Rangarajan,et al.  Transcription factors that mediate epithelial–mesenchymal transition lead to multidrug resistance by upregulating ABC transporters , 2011, Cell Death and Disease.

[148]  Steven I Hajdu,et al.  A note from history: Landmarks in history of cancer, part 2 , 2011, Cancer.

[149]  C. Sonnenschein,et al.  The tissue organization field theory of cancer: A testable replacement for the somatic mutation theory , 2011, BioEssays : news and reviews in molecular, cellular and developmental biology.

[150]  Hans Clevers,et al.  The cancer stem cell: premises, promises and challenges , 2011, Nature Medicine.

[151]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.

[152]  Hiroyuki Tomita,et al.  Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. , 2011, Cancer cell.

[153]  F. Sher,et al.  Ezh2 expression in astrocytes induces their dedifferentiation toward neural stem cells. , 2011, Cellular reprogramming.

[154]  Mauro Biffoni,et al.  Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells , 2011, Nature.

[155]  Rong Wang,et al.  Glioblastoma stem-like cells give rise to tumour endothelium , 2010, Nature.

[156]  V. Saradhi,et al.  Teratomas in central nervous system: a clinico-morphological study with review of literature. , 2010, Neurology India.

[157]  Jing Wang,et al.  Overexpression of EZH2 contributes to acquired cisplatin resistance in ovarian cancer cells in vitro and in vivo , 2010, Cancer biology & therapy.

[158]  J. Settleman,et al.  EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer , 2010, Oncogene.

[159]  T. Keck,et al.  ZEB1 in Pancreatic Cancer , 2010, Cancers.

[160]  P. Collodi,et al.  Zebrafish dead end possesses ATPase activity that is required for primordial germ cell development , 2010, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[161]  N. Ferrara,et al.  Targeting the tumour vasculature: insights from physiological angiogenesis , 2010, Nature Reviews Cancer.

[162]  Sven Christian,et al.  Origin and function of tumor stroma fibroblasts. , 2010, Seminars in cell & developmental biology.

[163]  K. Takeshita,et al.  Carcinosarcoma of the liver. , 2010, Internal medicine.

[164]  Miguel C. Seabra,et al.  Rab27a and Rab27b control different steps of the exosome secretion pathway , 2010, Nature Cell Biology.

[165]  E. D. De Robertis,et al.  Spemann’s organizer and the self-regulation of embryonic fields , 2009, Mechanisms of Development.

[166]  A. Ghanate,et al.  Snail and Slug Mediate Radioresistance and Chemoresistance by Antagonizing p53‐Mediated Apoptosis and Acquiring a Stem‐Like Phenotype in Ovarian Cancer Cells , 2009, Stem cells.

[167]  C. Mullighan,et al.  Human Gastrointestinal Neoplasia‐Associated Myofibroblasts Can Develop from Bone Marrow‐Derived Cells Following Allogeneic Stem Cell Transplantation , 2009, Stem cells.

[168]  R. Harland Induction into the Hall of Fame: tracing the lineage of Spemann's organizer , 2008, Development.

[169]  M. Todaro,et al.  Single-cell cloning of colon cancer stem cells reveals a multi-lineage differentiation capacity , 2008, Proceedings of the National Academy of Sciences.

[170]  M. Singhal,et al.  Primary mediastinal giant teratocarcinoma. , 2008, Indian journal of cancer.

[171]  Mary J. C. Hendrix,et al.  Reprogramming metastatic tumour cells with embryonic microenvironments , 2007, Nature Reviews Cancer.

[172]  J. Odorico,et al.  Undifferentiated murine embryonic stem cells cannot induce portal tolerance but may possess immune privilege secondary to reduced major histocompatibility complex antigen expression. , 2006, Stem cells and development.

[173]  A. Fritz,et al.  Zebrafish msxB, msxC and msxE function together to refine the neural-nonneural border and regulate cranial placodes and neural crest development. , 2006, Developmental biology.

[174]  Raghu Kalluri,et al.  Fibroblasts in cancer , 2006, Nature Reviews Cancer.

[175]  D. Solter,et al.  From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research , 2006, Nature Reviews Genetics.

[176]  E. D. Robertis,et al.  Spemann's organizer and self-regulation in amphibian embryos , 2006, Nature Reviews Molecular Cell Biology.

[177]  T. Curran,et al.  BGEM: An In Situ Hybridization Database of Gene Expression in the Embryonic and Adult Mouse Nervous System , 2006, PLoS biology.

[178]  M. Hendrix,et al.  Reprogramming metastatic melanoma cells to assume a neural crest cell-like phenotype in an embryonic microenvironment. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[179]  N. Benvenisty,et al.  Human Embryonic Stem Cells and Their Differentiated Derivatives Are Less Susceptible to Immune Rejection Than Adult Cells , 2006, Stem cells.

[180]  Chenbei Chang,et al.  Regulation of early Xenopus development by ErbB signaling , 2006, Developmental dynamics : an official publication of the American Association of Anatomists.

[181]  D. van der Kooy,et al.  Embryonic stem cells assume a primitive neural stem cell fate in the absence of extrinsic influences , 2006, The Journal of cell biology.

[182]  J. Nadeau,et al.  The Ter mutation in the dead end gene causes germ cell loss and testicular germ cell tumours , 2005, Nature.

[183]  E. D. De Robertis,et al.  Default neural induction: neuralization of dissociated Xenopus cells is mediated by Ras/MAPK activation. , 2005, Genes & development.

[184]  H. Spemann,et al.  über Induktion von Embryonalanlagen durch Implantation artfremder Organisatoren , 1924, Archiv für mikroskopische Anatomie und Entwicklungsmechanik.

[185]  Stuart R Hameroff,et al.  A new theory of the origin of cancer: quantum coherent entanglement, centrioles, mitosis, and differentiation. , 2004, Bio Systems.

[186]  E. D. De Robertis,et al.  Dorsal-ventral patterning and neural induction in Xenopus embryos. , 2004, Annual review of cell and developmental biology.

[187]  Rudolf Jaenisch,et al.  Reprogramming of a melanoma genome by nuclear transplantation. , 2004, Genes & development.

[188]  P. Johnstone,et al.  Primary retroperitoneal teratomas: A review of the literature , 2004, Journal of surgical oncology.

[189]  K. K. Chao,et al.  Sinonasal teratocarcinosarcoma. , 2004, American journal of clinical oncology.

[190]  J. Nichols,et al.  BMP Induction of Id Proteins Suppresses Differentiation and Sustains Embryonic Stem Cell Self-Renewal in Collaboration with STAT3 , 2003, Cell.

[191]  C. Uyttenhove,et al.  Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase , 2003, Nature Medicine.

[192]  T. Curran,et al.  Mouse embryos cloned from brain tumors. , 2003, Cancer research.

[193]  Z. Kozmík,et al.  Gelsolin is a dorsalizing factor in zebrafish , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[194]  Austin G Smith,et al.  Conversion of embryonic stem cells into neuroectodermal precursors in adherent monoculture , 2003, Nature Biotechnology.

[195]  J. Brickman,et al.  Pluripotency and tumorigenicity , 2002, Nature Genetics.

[196]  A. Knecht,et al.  Induction of the neural crest: a multigene process , 2002, Nature Reviews Genetics.

[197]  P. Andrews,et al.  From teratocarcinomas to embryonic stem cells. , 2002, Philosophical transactions of the Royal Society of London. Series B, Biological sciences.

[198]  Ali H. Brivanlou,et al.  Neural induction, the default model and embryonic stem cells , 2002, Nature Reviews Neuroscience.

[199]  R. Harland,et al.  The neural plate specifies somite size in the Xenopus laevis gastrula. , 2001, Developmental cell.

[200]  Janet Rossant,et al.  Direct Neural Fate Specification from Embryonic Stem Cells A Primitive Mammalian Neural Stem Cell Stage Acquired through a Default Mechanism , 2001, Neuron.

[201]  A. Otte,et al.  The Polycomb Group Protein EED Interacts with YY1, and Both Proteins Induce Neural Tissue in XenopusEmbryos , 2001, Molecular and Cellular Biology.

[202]  J. Gerhart,et al.  Evolution of the organizer and the chordate body plan. , 2001, The International journal of developmental biology.

[203]  H. Spemann,et al.  Induction of Embryonic Primordia by Implantation of Organizers from a Different Species. , 2024, Cells & development.

[204]  U. Lendahl,et al.  Generalized potential of adult neural stem cells. , 2000, Science.

[205]  S. Gilbert Neural crest cells and axonal specificity , 2000 .

[206]  R. Harland Neural induction. , 2000, Current opinion in genetics & development.

[207]  Yang Shi,et al.  Targeted Disruption of Mouse Yin Yang 1 Transcription Factor Results in Peri-Implantation Lethality , 1999, Molecular and Cellular Biology.

[208]  M. Levin Twinning and embryonic left-right asymmetry. , 1999, Laterality.

[209]  D. Riethmacher,et al.  The ErbB2 and ErbB3 receptors and their ligand, neuregulin-1, are essential for development of the sympathetic nervous system. , 1998, Genes & development.

[210]  D. Gottlieb,et al.  Neural cells derived by in vitro differentiation of P19 and embryonic stem cells. , 1998, Perspectives on developmental neurobiology.

[211]  J. Sleeman,et al.  Autonomous neural axis formation by ectopic expression of the protooncogene c-ski. , 1997, Developmental biology.

[212]  J. J. Hoorweg,et al.  Osteoid and bone formation in a nasal mucosal melanoma and its metastasis , 1997, Histopathology.

[213]  C. Colmenares,et al.  Mice lacking the ski proto-oncogene have defects in neurulation, craniofacial, patterning, and skeletal muscle development. , 1997, Genes & development.

[214]  A. Hemmati‐Brivanlou,et al.  Neural induction in Xenopus laevis: evidence for the default model , 1997, Current Opinion in Neurobiology.

[215]  T. Bouwmeester,et al.  Cerberus is a head-inducing secreted factor expressed in the anterior endoderm of Spemann's organizer , 1996, Nature.

[216]  D. Saranath,et al.  A neural precursor cell line derived from murine teratocarcinoma. , 1996, The International journal of developmental biology.

[217]  M J Bissell,et al.  Cellular changes involved in conversion of normal to malignant breast: importance of the stromal reaction. , 1996, Physiological reviews.

[218]  Rüdiger Klein,et al.  Aberrant neural and cardiac development in mice lacking the ErbB4 neuregulin receptor , 1995, Nature.

[219]  K. Herrup,et al.  Targeted disruption of mouse EGF receptor: effect of genetic background on mutant phenotype. , 1995, Science.

[220]  M. Bronner‐Fraser,et al.  Origins of the avian neural crest: the role of neural plate-epidermal interactions. , 1995, Development.

[221]  T. Magnuson,et al.  The eed mutation disrupts anterior mesoderm production in mice. , 1995, Development.

[222]  Y. Sasai,et al.  Xenopus chordin: A novel dorsalizing factor activated by organizer-specific homeobox genes , 1994, Cell.

[223]  D. Melton,et al.  Inhibition of activin receptor signaling promotes neuralization in Xenopus , 1994, Cell.

[224]  Virginia M. Y. Lee,et al.  NTera 2 Cells: A human cell line which displays characteristics expected of a human committed neuronal progenitor cell , 1993, Journal of neuroscience research.

[225]  K. Kinzler,et al.  The multistep nature of cancer. , 1993, Trends in genetics : TIG.

[226]  William C. Smith,et al.  Expression cloning of noggin, a new dorsalizing factor localized to the Spemann organizer in Xenopus embryos , 1992, Cell.

[227]  L. Tacke,et al.  Neural differentiation of Xenopus laevis ectoderm takes place after disaggregation and delayed reaggregation without inducer. , 1989, Cell differentiation and development : the official journal of the International Society of Developmental Biologists.

[228]  J. Slack,et al.  Clonal analysis of mesoderm induction in Xenopus laevis. , 1989, Developmental biology.

[229]  T. Sargent,et al.  Development of neural inducing capacity in dissociated Xenopus embryos. , 1989, Developmental biology.

[230]  Zhen-yi Wang,et al.  Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia. , 1988, Haematology and blood transfusion.

[231]  S D Carson,et al.  Regulation of melanoma by the embryonic skin. , 1986, Proceedings of the National Academy of Sciences of the United States of America.

[232]  R. S. Wells,et al.  Widespread inhibition of neuroblastoma cells in the 13- to 17-day-old mouse embryo. , 1986, Cancer research.

[233]  H. Rubin Cancer as a dynamic developmental disorder. , 1985, Cancer research.

[234]  M. Moore,et al.  Human tumor-infiltrating lymphocytes: a marker of host response. , 1985, Seminars in hematology.

[235]  G. B. Pierce,et al.  The neurula stage mouse embryo in control of neuroblastoma. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[236]  L. Sachs,et al.  Developmental potential of myeloid leukemia cells injected into midgestation embryos. , 1984, Developmental biology.

[237]  S. Kottaridis,et al.  Uncontrolled growth of tumour stromal fibroblasts in vitro. , 1983, Experimental cell biology.

[238]  M. Mizell,et al.  Frog larvae cloned from nuclei of pronephric adenocarcinoma. , 1982, Differentiation; research in biological diversity.

[239]  G. B. Pierce The cancer cell and its control by the embryo. Rous-Whipple Award lecture. , 1983, The American journal of pathology.

[240]  L. Sachs,et al.  Participation of myeloid leukaemic cells injected into embryos in haematopoietic differentiation in adult mice , 1982, Nature.

[241]  H. Macdonald,et al.  Quantitation and clonal isolation of cytolytic T lymphocyte precursors selectively infiltrating murine sarcoma virus-induced tumors , 1981, The Journal of experimental medicine.

[242]  S. Argov,et al.  Natural cytotoxicity in man: activity of lymph node and tumor‐infiltrating lymphocytes , 1977, European journal of immunology.

[243]  H. Pinkus,et al.  Intrauterine transplantation of rat basal cell carcinoma as a model for reconversion of malignant to benign growth. , 1977, Cancer research.

[244]  K. Illmensee,et al.  Totipotency and normal differentiation of single teratocarcinoma cells cloned by injection into blastocysts. , 1976, Proceedings of the National Academy of Sciences of the United States of America.

[245]  M. Evans,et al.  Fate of teratocarcinoma cells injected into early mouse embryos , 1975, Nature.

[246]  R. Brinster THE EFFECT OF CELLS TRANSFERRED INTO THE MOUSE BLASTOCYST ON SUBSEQUENT DEVELOPMENT , 1974, The Journal of experimental medicine.

[247]  G. B. Pierce,et al.  Differentiation of malignant to benign cells. , 1971, Cancer research.

[248]  R. McKinnell,et al.  Transplantation of Pluripotential Nuclei from Triploid Frog Tumors , 1969, Science.

[249]  T. J. King,et al.  TRANSPLANTATION OF NUCLEI FROM THE FROG RENAL ADENOCARCINOMA I. DEVELOPMENT OF TUMOR NUCLEAR‐TRANSPLANT EMBRYOS * , 1965, Annals of the New York Academy of Sciences.

[250]  R. R. Bensley,et al.  Embryonic Development and Induction , 1938, The Yale Journal of Biology and Medicine.