BC Basics 2017.

Slides:



Advertisements
Similar presentations
Cancer Genetics Is Cancer a Genetic Disease? Cancer is not a classic genetic disease, instead, Genetic background (set-up) has a definite role in cancer.
Advertisements

Lecture 11: Signalling for Life/Death 1)Describe the eukaryotic cell cycle and the purpose of checkpoints. 2)Describe the role of cyclins and cyclin-dependent.
BioSci 145A lecture 18 page 1 © copyright Bruce Blumberg All rights reserved BioSci 145A Lecture 18 - Oncogenes and Cancer Topics we will cover today.
21 and 23 March, 2005 Chapter 15 Regulation of Cell Number: Normal and Cancer Cells Regulated and unregulated cell proliferation.
BRCA Genes Dallas Henson.
Chap. 16 Signaling Pathways That Control Gene Expression
Manifestation of Novel Social Challenges of the European Union in the Teaching Material of Medical Biotechnology Master’s Programmes at the University.
Signal Transduction Biochemistry – February 23, 2005 Chapter 12 – parts 12.3, 12.4.
Cell Communication Chapter 9. Please note that due to differing operating systems, some animations will not appear until the presentation is viewed in.
Cancer and the Cell Cycle. Outline of the lecture n What is cancer? n Review of the cell cycle and regulation of cell growth n Which types of genes when.
Cell Signaling II Signal Transduction pathways
 Regulation of Cell Number and Cancer Cells Special Limited Edition Packet Tuesday, November 10,
Benign Versus Malignant Tumors
Biochemistry Sixth Edition Chapter 31 The Control of Gene Expression Part II: Eukaryotes (cis vs. trans) Copyright © 2007 by W. H. Freeman and Company.
Specialized signaling pathways 1: RTK associated pathways
 Signaling molecules that function within an organism to control metabolic processes within cells, the growth and differentiation of tissues, the synthesis.
EUKARYOTIC CELL SIGNALING VII Abnormal Signaling in Cancer Signaling to p53 Dr. Ke Shuai Office: 9-240M Factor Tel: X69168
Protein Receptors & Signal Transduction
Epidermal growth factor receptor tyrosine kinase inhibitors as initial therapy for non- small cell lung cancer: Focus on epidermal growth factor receptor.
Cell Signaling Pathways – A Case Study Approach
Dietary cancer-chemopreventive compounds: from signaling and gene expression to pharmacological effects  Chi Chen, Ah-Ng Tony Kong  Trends in Pharmacological.
REGULATION OF GENE EXPRESSION
Figure 1. Resistance mechanism against first generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI). (A) Mutations in the EGFR.
The nature, significance, and glucagon-like peptide-1 analog treatment of brain insulin resistance in Alzheimer's disease  Konrad Talbot, Hoau-Yan Wang 
Med Princ Pract 2016;25(suppl 2): DOI: /
Controls the Cell Cycle
Regulation of Gene Expression
Cellular and Molecular Mechanisms of Liver Injury
Concept 18.5: Cancer results from genetic changes that affect cell cycle control The gene regulation systems that go wrong during cancer are the very same.
Sustaining Proliferative Signaling and Evading Growth Suppressors
Chap. 16 Problem 1 Cytokine receptors and RTKs both form functional dimers on binding of ligand. Ligand binding activates cytosolic kinase domains which.
Ian M. Adcock, PhD, Kittipong Maneechotesuwan, MD, Omar Usmani, MBBS 
You have identified a novel cytoplasmic protein
Genetics Of Cancer Regulation of cell proliferation and cancer
B lymphocytes produce antibodies.
PTEN (a.k.a. MMAC1 and TEP1) and Cowden’s Disease
Cellular and Molecular Mechanisms of Liver Injury
Figure 1 mTOR pathway activation
Regulation of Gene Expression
Figure 1 A schematic representation of the HER2 signalling pathway
Figure 2 Oestrogen receptor signalling pathways
Figure 3 Physiological regulation of autophagy in the heart
BDNF and insulin signaling pathways activated by phytochemicals.
Nat. Rev. Nephrol. doi: /nrneph
Figure 1 mTOR complex biology
Selective estrogen receptor modulation
Volume 29, Issue 3, Pages (March 2016)
PTEN Tumor Suppressor and Cancer
The pathway signalling starts with the binding of insulin or growth factors to insulin receptors. The pathway signalling starts with the binding of insulin.
Development of PI3K/AKT/mTOR Pathway Inhibitors and Their Application in Personalized Therapy for Non–Small-Cell Lung Cancer  Vassiliki Papadimitrakopoulou,
Jing Zhang, Thomas M. Roberts, Ramesh A. Shivdasani  Gastroenterology 
Bhagelu Ram Achyut, Li Yang  Gastroenterology 
Endocrinology Introduction Lecture 3.
Unit III Information Essential to Life Processes
Figure 1 The insulin signalling pathway
Taru Muranen, Funda Meric-Bernstam, Gordon B. Mills  Cancer Cell 
FGFR Signaling as a Target for Lung Cancer Therapy
An Emerging Role of mTOR in Lipid Biosynthesis
mTORC1 Signaling: A Double-Edged Sword in Diabetic β Cells
AKT/PKB Signaling: Navigating the Network
AKT/PKB Signaling: Navigating Downstream
Signalling links in the liver: Knitting SOCS with fat and inflammation
The nature, significance, and glucagon-like peptide-1 analog treatment of brain insulin resistance in Alzheimer's disease  Konrad Talbot, Hoau-Yan Wang 
Volume 104, Issue 4, Pages (February 2001)
Dysregulation of the mTOR Pathway Secondary to Mutations or a Hostile Microenvironment Contributes to Cancer and Poor Wound Healing  Richard A.F. Clark,
Nutrient Sensing, Metabolism, and Cell Growth Control
Brief Review – Growth Factors and Receptors
Cell Signaling by Receptor Tyrosine Kinases
Successful targeting of ErbB2 receptors—is PTEN the key?
Presentation transcript:

BC Basics 2017

BC as a heterogeneous group of disorders Incidence- 1:9 Multi- stage process (initiation, promotion)

Heterogeneous approaches for diagnosis Triple test (imaging, FNA, examination) Sub- type: Immunostaining Genomics

ER receptor signaling enhancement in BC

ER signaling cascade According to the “canonical model”, following E2-binding, ER dimers bind to specific 13-bp DNA sequences in the estrogen receptor response element ERE characteristic of specific E2-regulated genes HSPs as quantity modulators

DNA bound ER-agonist complexes recruit several coactivators some with histone acetyl transferase (HAT) activities that allow repositioning of nucleosome and chromatin opening and interaction with the general transcription machinery organized around RNA polymerase.

What happens if and when the number of ERs is increased in a cell? b: ER activation by RTKs (EGFR, ERBB2, IGFR). Phosphorylation (P) by the Erk or Akt serine/threonine kinases leads to ligand-independent activation of the ER. formation of an ER–PI3K–Src–focal adhesion kinase (FAK) complex that activates Akt (d), and activation of Erk by ER–Src–PELP1 complexes (e) What happens if and when the number of ERs is increased in a cell?

Treatment Selective estrogen-receptor response modulators- TAMOXIPHEN Estrogen-receptor down -regulators (3rd line) Aromatase inhibitors- inhibition of aromatase inhibit the production of E2 in fat tissue and adrenal (PM women) Prophylactic Ovaries removal Similar approaches for PR

Growth factors response Convergence of 2 major cascades Promotes cell cycle, cell sustainability ERB2 participate in signaling w/o agonist Amplification of ERB2/ HER2 20% of breast cancers, poor prognosis

Directed/ tailored therapies Trastuzumab (Herceptin): a humanized monoclonal antibody targeted against the extracellular portion of HER2 Mechanism of action Inhibition of Cyclin D Reducing GF paracrine secretion Inducing immunologic response (?) Early diagnosis – improved prognosis Late diagnosis- resistance, Always small molecules TKI- Lapatinib (Glaxo) Oral, CYPD metabolism EGFR and HER competitive inhibitor Usually in combination for advanced cancers

Resistance Look at PTEN Activating mutations in each signaling cascade Increased HER2-HER3 heterodimers Changing the HER1 structure Other routes of signaling P53 New approach- combination therapies New improved formulations/ structures TKI-Herceptin MTORE inhibitors

Oestrogen-bound oestrogen receptor (ER), in association with a variety of co-activators (CoAs) (part a) and co-repressors (CoRs) (part b), exerts its classical genomic action as a transcription factor through the oestrogen response element (ERE) of target genes. ER can also mediate an ERE-independent genomic effect via interaction with other transcription factors, such as AP1, through AP1-binding sites of target genes (part c). In addition, ER can be activated via plasma membrane crosstalk with other growth factor receptor (GFR) pathways that phosphorylate (P) ER or its co-regulators. GFRs — for example, HER2, epidermal growth factor receptor (EGFR), insulin-like growth factor 1 receptor (IGF1R) and fibroblast growth factor receptor (FGFR) — undergo phosphorylation and dimerization upon ligand activation, which triggers their intracellular association with the regulatory subunit (p85) of PI3K, either directly or indirectly via an intermediate adaptor molecule such as insulin receptor substrate 1 (IRS1). This releases the inhibitory effect of p85 on the catalytic subunit p110 of PI3K. PI3K converts phosphatidylinositol bisphosphate (PIP2) to phosphatidylinositol trisphosphate (PIP3), which allows the activation of AKT and downstream signalling components of the PI3K pathway to promote cell growth, proliferation and survival. PTEN and inositol polyphosphate 4-phosphatase type II (INPP4B) are two phosphatases that negatively regulate the PI3K pathway by dephosphorylating PIP3 and PIP2, respectively. In addition, activation of receptor tyrosine kinases (RTKs) also activates the MAPK pathway. PI3K and MAPK pathway activation leads to phosphorylation of ER and ER coregulators. Cyclin D1 is a direct transcription target of ER or other GFR signalling pathways. Cyclin D1 activates cyclin-dependent kinase 4 (CDK4) and CDK6, which phosphorylates RB, which releases the E2F transcription factors and activates the expression of genes required for the G1 to S phase transition of the cell cycle. CDK4 and CDK6 are negatively regulated by specific and universal inhibitors including p16, p18, p21 and p27. p53 negatively regulates the cell cycle through the activation of p21. MDM2 inhibits p53 function. RB and p53 are important mediators of apoptosis and/or senescence with intrinsic or extrinsic signals such as genotoxic stress. The crosstalk between ER and GFR signalling, as well as constitutive activation of their downstream effectors, are potential mechanisms of oestrogen-independent cell growth of ER+ breast cancer, leading to aromatase inhibitor (AI) resistance. 4EBP1, eukaryotic translation initiation factor 4E-binding protein 1; AMPK, AMP-activated protein kinase; ASK1, apoptosis signal-regulating kinase 1; BAD, BCL-2-associated agonist of cell death; eIF4, eukaryotic translation initiation factor 4; FOXO, forkhead box protein O; GRB2, growth factor receptor-bound protein 2; GSK3, glycogen synthase kinase 3; JNK, JUN N-terminal kinase; LKB1, liver kinase B1; mTORC, mTOR complex; PDK1, 3-phosphoinositide-dependent protein kinase 1; RHEB, RAS homologue enriched in brain; SHIP, SH2 domain-containing inositol 5′ phosphatase.

Others – BRCA genes

Incidence Two genes predispose to BC if they are mutated* General population carriers: 0.2 to 0.3% Ashkenazi Jewish population: 2.5% Women with breast cancer: 3% Women with breast cancer onset before age 40 years: 6% up Women with ovarian cancer: 10% up High-risk families (higher incidence of BC and other C) : 20% A mutation in those genes confers a high risk to BC the early age of onset (+ other findings) – inherited risk= the greater risk ovarian, pancreatic, stomach, laryngeal, fallopian tube and prostate cancer

Genetics One mutated allele in the germline= hereditary risk Both are tumor suppressors- “haplo” is not a problem 2nd hit model of inherited cancers “pushing” to cellular tumor initiation (LOH) Both proteins participate in homologous recombination during repair of DSB (error safe) DNA Damage Response (DDR) elicits and depends on signaling cascades with multiple components Control check-points Genome integrity Replication fork faliure

BRCA2: Recruits recombinase RAD51 and binds to DNA (ss, ds) BRCA1: Connects to the damage sensing molecules and recruits effectors through multiple domains x8 repeats! BRCA2: Recruits recombinase RAD51 and binds to DNA (ss, ds)

Protein Ubi (by ring), chromatin change by H2X P, Enzymes-RAD, Recognition by ATM/ATR

So why BC? Reduced HR repair capacity leading to error prone mechanisms Malfunctioning off other tumor suppressors (P53, ATM) Why in Breast and Ovarian tissues? Sporadic cases: probably due to HR reduced efficiency but via mutations in other DDR participants.