molecular basis of cancer part 1

41
PART 1 DR. GANGADHAR CHATTERJEE MOLECULAR BASIS OF CANCER

Upload: gangadhar-chatterjee

Post on 18-Dec-2014

1.138 views

Category:

Health & Medicine


4 download

DESCRIPTION

 

TRANSCRIPT

Page 1: Molecular basis of cancer part 1

PART 1DR. GANGADHAR CHATTERJEE

MOLECULAR BASIS OF CANCER

Page 2: Molecular basis of cancer part 1

Neoplasia means “new growth”

Greek oncos = tumor British oncologist Willis gave closest

definition-“A neoplasm is an abnormal mass of tissue, the growth of which exceeds and is uncoordinated with that of the normal tissues and persists in the same excessive manner after cessation of the stimuli which evoked the change.”

Malignant tumors are collectively referred to as cancers, derived from the Latin word for crab

Page 3: Molecular basis of cancer part 1

All tumors, benign and malignant, have two basic components

clonal neoplastic cells that constitute their parenchyma

reactive stroma made up of connective tissue, blood vessels, and variable numbers of macrophages and lymphocytes

DIFFERENTIATION AND ANAPLASIA

Differentiation- the extent to which neoplastic parenchymal cells resemble the corresponding normal parenchymal cells, both morphologically and functionally;

lack of differentiation called anaplasia

In general, benign tumors are well differentiated

Malignant neoplasms - wide range of parenchymal cell differentiation, from surprisingly well differentiated to completely undifferentiated

Page 4: Molecular basis of cancer part 1

METASTASIS

tumor implants discontinuous with the primary tumor

unequivocally marks a tumor as malignant because benign neoplasms do not metastasize

Characteristics Benign MalignantDifferentiation/anaplasia Well differentiated;

structure sometimes typical of tissue of origin

Some lack of differentiation with anaplasia; structure often atypical

Rate of growth Usually progressive and slow; may come to a standstill or regress; mitotic figures rare and normal

Erratic and may be slow to rapid; mitotic figures may be numerous and abnormal

Local invasion Usually cohesive expansile well-demarcated masses that do not invade or infiltrate surrounding normal tissues

Locally invasive, infiltrating surrounding tissue; sometimes may be seemingly cohesive and expansile

Metastasis Absent Frequently present; the larger and more undifferentiated the primary, the more likely are metastases

Comparisons between Benign and Malignant Tumors

Page 5: Molecular basis of cancer part 1

5

What causes Cancer? Cancer is caused by

alterations or mutations in the genetic code

Can be induced in somatic cells by:

Carcinogenic

chemicals Radiation Some viruses

Heredity - 5%

Page 6: Molecular basis of cancer part 1

INHERITED CANCER SYNDROMES (AUTOSOMAL DOMINANT)Gene Inherited PredispositionRB Retinoblastomap53 Li-Fraumeni syndrome (various tumors)

p16/INK4A MelanomaAPC Familial adenomatous polyposis/colon cancer

NF1, NF2 Neurofibromatosis 1 and 2BRCA1, BRCA2 Breast and ovarian tumorsMEN1, RET Multiple endocrine neoplasia 1 and 2

MSH2, MLH1, MSH6 Hereditary nonpolyposis colon cancer

PTCH Nevoid basal cell carcinoma syndrome

PTEN Cowden syndrome (epithelial cancers)

LKB1 Peutz-Jegher syndrome (epithelial cancers)

VHL Renal cell carcinomasINHERITED AUTOSOMAL RECESSIVE SYNDROMES OF DEFECTIVE DNA REPAIR

Xeroderma pigmentosumAtaxia-telangiectasiaBloom syndromeFanconi anemiaFAMILIAL CANCERSFamilial clustering of cases, but role of inherited predisposition not clear for each individual

 Breast cancer Ovarian cancer Pancreatic cancer

Examples of Inherited Predisposition to Cancer

Page 7: Molecular basis of cancer part 1

Molecular Basis of Cancer

Page 8: Molecular basis of cancer part 1

Nonlethal genetic damage lies at the heart of carcinogenesis

A tumor is formed by the clonal expansion of a single precursor cell that has incurred genetic damage (i.e., tumors are monoclonal).

most commonly used method to determine tumor clonality involves the analysis of methylation patterns adjacent to the highly polymorphic locus of the human androgen receptor gene, AR

Four classes of normal regulatory genes—the growth-promoting proto-oncogenes, the growth-inhibiting tumor suppressor genes, genes that regulate programmed cell death (apoptosis), and genes involved in DNA repair-are the principal targets of genetic damage

Carcinogenesis is a multistep process at both the phenotypic and the genetic levels, resulting from the accumulation of multiple mutations

Page 9: Molecular basis of cancer part 1

Even though most malignant tumors are monoclonal in origin, by the time they become clinically evident their constituent cells are extremely heterogeneous.

Also they will undergo immune surveillance.

Tumor progression and generation of heterogeneity.

Page 10: Molecular basis of cancer part 1

ESSENTIAL ALTERATIONS FOR MALIGNANT TRANSFORMATION

Page 11: Molecular basis of cancer part 1

Flowchart depicting a simplified scheme of the molecular basis of cancer

Page 12: Molecular basis of cancer part 1

SELF-SUFFICIENCY IN GROWTH SIGNALS: ONCOGENES

Genes that promote autonomous cell growth in cancer cells are called oncogenes.

unmutated cellular counterparts are called proto-oncogenes.

Products of oncogene called oncoproteins

sequential steps that characterize normal cell proliferation

• The binding of a growth factor to its specific receptor

• Transient and limited activation of the growth factor receptor, which, in turn, activates several signal-transducing proteins on the inner leaflet of the plasma membrane

• Transmission of the transduced signal across the cytosol to the nucleus via second messengers or by a cascade of signal transduction molecules

• Induction and activation of nuclear regulatory factors that initiate DNA transcription

• Entry and progression of the cell into the cell cycle, ultimately resulting in cell division

Page 13: Molecular basis of cancer part 1

Selected Oncogenes, Their Mode of Activation, and Associated Human Tumors

Page 14: Molecular basis of cancer part 1

Selected Oncogenes, Their Mode of Activation, and Associated Human

Tumors

Page 15: Molecular basis of cancer part 1

Growth Factors.

- switching from normal paracrine loop to autocrine loop.

-e.g. PDGF in many glioblastomas, TGF-α

Growth Factor Receptors.

-transmembrane proteins with an external ligand-binding domain and a cytoplasmic tyrosine kinase domain

-the kinase is transiently activated by binding of the specific growth factors, followed rapidly by receptor dimerization and tyrosine phosphorylation of several substrates switching on the signaling cascade.

The oncogenic versions of these receptors are associated with constitutive dimerization and activation without binding to the growth factor.

Page 16: Molecular basis of cancer part 1

RET protooncogene, a receptor tyrosine kinase, exemplifies

oncogenic conversion via mutations and gene rearrangements RET protein- receptor for the glial cell line–derived

neurotrophic factor

structurally related proteins that promote cell survival during neural development

normally expressed in neuroendocrine cells, such as parafollicular C cells of the thyroid, adrenal medulla, and parathyroid cell precursors

Point mutations in the RET proto-oncogene are associated with dominantly inherited MEN types 2A and 2B and familial medullary thyroid carcinoma

Other example- FLT-3 (FMS like Tyrosine Kinase3) mutation in myeloid leukaemia.

> 90% of GIST-mutation in the receptor tyrosine kinase c-KIT or

PDGFR. Imatinib mesylate is the T/t – a tyrosine kinase inhibitor.

Page 17: Molecular basis of cancer part 1

more common than mutations is overexpression of normal forms of

growth factor receptor Two members of the epidermal growth factor (EGF)

receptor family best described

normal form of ERBB1, the EGF receptor gene, overexpressed in up to 80% of squamous cell carcinomas of the lung, in 50% or more of glioblastomas and in 80% to 100% of head and neck tumors

ERBB2 gene (also called HER-2/NEU), the second member of the EGF receptor family, amplified in approximately 25% of breast cancers and in human adenocarcinomas arising within the ovary, lung, stomach, and salivary glands

Page 18: Molecular basis of cancer part 1

Signal-Transducing Proteins. most well-studied example of a signal transducing

oncoprotein is the RAS family of guanine triphosphate (GTP)-binding proteins (G proteins)

The RAS Oncogene

-three types in human genome (HRAS, KRAS, NRAS)

-Point mutation of RAS family genes is the single most common abnormality of proto-oncogenes in human tumors.

-carcinomas (particularly from colon and pancreas)

have mutations of KRAS, bladder tumors have HRAS mutations, and hematopoietic tumors bear NRAS mutations.

RAS plays an important role in signaling cascades downstream of growth factor receptors, resulting in mitogenesis.

Page 19: Molecular basis of cancer part 1

Model for action of RAS genes.

Page 20: Molecular basis of cancer part 1

Transcription Factors Transcription factors contain specific amino acid sequences or

motifs that allow them to bind DNA or to dimerize for DNA binding

A host of oncoproteins, including products of the MYC, MYB, JUN, FOS, and REL oncogenes, are transcription factors that regulate the expression of growth-promoting genes, such as cyclins.

MYC is most commonly involved in human tumors

The MYC Oncogene

expressed in virtually all eukaryotic cells

belongs to the immediate early response genes, which rapidly induced when quiescent cells receive a signal to divide

the range of activities modulated by MYC very broad

includes histone acetylation, reduced cell adhesion, increased cell motility, increased telomerase activity, increased protein synthesis, decreased proteinase activity

one of a handful of transcription factors that can act in concert to reprogram somatic cells into pluripotent stem cells

Page 21: Molecular basis of cancer part 1

Cyclins and Cyclin-Dependent Kinases

ultimate outcome of all growth-promoting stimuli is the entry of quiescent cells into the cell cycle.

orderly progression of cells through the various phases of the cell cycle is orchestrated by cyclin-dependent kinases (CDKs)

CDKs activated by binding to cyclins, so called because of the cyclic nature of their production and degradation

The CDK-cyclin complexes phosphorylate crucial target proteins that drive the cell through the cell cycle

cyclins D, E, A, and B appear sequentially during the cell cycle and bind to one or more CDK

Page 22: Molecular basis of cancer part 1

Schematic illustration of the role of cyclins, CDKs, and CDK inhibitors (CDKIs) in regulating the cell

cycle

Page 23: Molecular basis of cancer part 1

Internal controls of the cell cycle(checkpoints)

G1/S transition

G2/M

S phase, the point of no return in the cell cycle

delay in cell cycle progression provides the time needed for DNA repair

if the damage not repairable, apoptotic pathways activated to kill the cell.

G2/M checkpoint monitors the completion of DNA replication, particularly important in cells exposed to ionizing radiation.

Defects in cell cycle checkpoint components, a major cause of genetic instability in cancer cells.

Page 24: Molecular basis of cancer part 1

INSENSITIVITY TO GROWTH INHIBITION AND ESCAPE FROM

SENESCENCE TUMOR SUPPRESSOR GENES

Page 25: Molecular basis of cancer part 1

Tumor suppressor genes apply brakes to cell proliferation

RB and p53, part of a regulatory network that recognizes genotoxic stress from any source, and responds by shutting down proliferation.

The RETINOBLASTOMA (RB) gene RB, the first, and prototypic, tumor

suppressor gene discovered.

Knudson’s canonical “two-hit” hypothesis of oncogenesis

Two mutations (hits), involving both alleles of RB at chromosome locus 13q14, are required to produce retinoblastoma.

Page 26: Molecular basis of cancer part 1

In familial cases, children inherit one defective copy of the RB gene in the germ line (one hit);

Retinoblastoma develops when the normal RB allele is mutated in retinoblasts as a result of spontaneous somatic mutation (second hit).

Loss of heterozygosity

Page 27: Molecular basis of cancer part 1

RB protein, the product of the RB gene.-ubiquitously expressed nuclear phosphoprotein-plays a key role in regulating the cell cycle.-RB exists in active hypophosphorylated state in quiescent cells-inactive hyperphosphorylated state in the G1/S cell cycle transition POLICING THE G1-S CHECK POINT

Page 28: Molecular basis of cancer part 1

p53: Guardian of the Genome located on chromosome 17p13.1

most common target for genetic alteration in human tumors.

A little over 50% of human tumors contain mutations in this gene.

Homozygous loss of p53 occurs in virtually every type of cancer, including carcinomas of the lung, colon, and breast—the three leading causes of cancer death

Mutation occurs usually somatically, not germline.

If occurs germline, only one HIT required for carcinogenesis – called Li-Fraumeni syndrome

P53, transcription factor that sense cellular stress, such as DNA damage, shortened telomeres, and hypoxia.

Page 29: Molecular basis of cancer part 1

p53 thwarts neoplastic transformation by three interlocking mechanisms: -activation of temporary cell cycle arrest (quiescence), - induction of permanent cell cycle arrest (senescence),-triggering of programmed cell death (apoptosis).

Page 30: Molecular basis of cancer part 1

-In nonstressed, healthy cells, p53 has a short half-life (20 minutes), because of its associationwith MDM2, a protein that targets it for destruction. When the cell is stressed, p53 undergoes post-

transcriptional modifications that release it from MDM2 and increase its half-life.

?? How p53 repress gene expression??

MicroRNA

Targets of mir34s include pro-proliferative genes such as cyclins, and anti-apoptotic genes such as BCL2.

p53 regulation of mir34 explains, at least in part, how p53 is able to repress gene expression, and it seems that regulation of this miRNA is crucial for the p53 response.

Page 31: Molecular basis of cancer part 1

Small guy with big clubs- the microRNA

blocking mir34 severely hampered the p53response in cells

Page 32: Molecular basis of cancer part 1

The key initiators of the DNA-damage pathway are two related protein kinases:

-ataxia-telangiectasia mutated (ATM)

-ataxia-telangiectasia and Rad3 related (ATR).

p53-mediated cell cycle arrest may be considered the primordial response to DNA damage

p21 inhibits cyclin-CDK complexes and phosphorylation of RB, thereby preventing cells from entering G1 phase

p53-induced senescence is a permanent cell cycle arrest

p53-induced apoptosis of cells with irreversible DNA damage is the ultimate protective mechanism against neoplastic transformation

Page 33: Molecular basis of cancer part 1

APC/β-Catenin Pathwaychromosome 5q21

APC, a component of the WNT signaling pathway, which has a major role in controlling cell fate, adhesion, and cell polarity during embryonic development.

WNT signaling is required for self-renewal of hematopoietic stem cells.

An important function of the APC protein is to down-regulate β-catenin. In the absence of WNT signaling APC causes degradation of β-catenin, preventing its accumulation in the cytoplasm.

mutations in the β-catenin gene are present in more than 50% of hepatoblastomas and in approximately 20% of hepatocellular carcinomas.

Page 34: Molecular basis of cancer part 1

The role of APC in regulating the stability and function of β-catenin.

Page 35: Molecular basis of cancer part 1

Other tumour suppressor genes INK4a/ARF

encodes two protein products-

1) p16/INK4a CDKI, which blocks cyclin D/CDK2-mediated phosphorylation of RB, keeping the RB

checkpoint in place.

2) p14/ARF, activates the p53 pathway by

inhibiting MDM2 and preventing destruction of p53.

p16 in particular is crucial for the induction of senescence.

Mutations at this locus detected in bladder, head and neck tumors, acute lymphoblastic leukemias, and

cholangiocarcinomas.

Page 36: Molecular basis of cancer part 1

The TGF-β Pathway

In most normal epithelial, endothelial, and hematopoietic cells, TGF-β is a potent inhibitor of proliferation

regulates cellular processes by binding to a serine-threonine kinase complex composed of TGF-β receptors I and II.

TGF-β signaling leads to repression of c-MYC, CDK2, CDK4, and cyclins A and E.

In 100% of pancreatic cancers and 83% of colon cancers, at least one component of the TGF-β pathway is mutated.

Page 37: Molecular basis of cancer part 1

PTEN (Phosphatase and tensin homologue)

a membrane-associated phosphatase encoded by a gene on chromosome 10q23

mutated in Cowden syndrome, an autosomal dominant disorder marked by frequent benign growths

acts as a tumor suppressor by serving as a brake on the prosurvival/ pro-growth PI3K/AKT pathway

this pathway normally stimulated (along with the RAS and JAK/STAT pathways) when ligands bind to receptor tyrosine kinases and involves a cascade of phosphorylation events.

Page 38: Molecular basis of cancer part 1

38 Revolution in cancer treatment: ‘Smart Bullets Period’

Page 39: Molecular basis of cancer part 1

Summary of 30 years of research (1971-2001)

Page 40: Molecular basis of cancer part 1

40

Bilimsel Araştırmaların Kanserle Savaşa KatkısıHERCEPTİN

HERCEPTIN

STI-571

Page 41: Molecular basis of cancer part 1

Thank you