B33E11 Complete TB

download B33E11 Complete TB

of 177

description

h

Transcript of B33E11 Complete TB

  • Cancer Biology and Chemotherapy

    Tracey D Bradshaw

    E-mail: [email protected]

    Outline:

    Introduction to cancer

    Cytotoxic and Target-directed cancer chemotherapy

  • Appreciate the size of the cancer problem and the challenge facing

    patients, clinicians and researchers

    Incidence

    Mortality

    Objectives (i)

  • Appreciate the size of the cancer problem and the challenge facing

    patients, clinicians and researchers

    Incidence

    Mortality

    Recognise common cancer terminology

    Apreciate some causes of cancer epidemiology

    Objectives (i)

  • Appreciate the size of the cancer problem and the challenge facing

    patients, clinicians and researchers

    Incidence

    Mortality

    Recognise common cancer terminology

    Apreciate some causes of cancer epidemiology

    Cancer is a genetic disease

    Oncogenes

    Tumour suppressor genes

    DNA repair genes/mechanisms gatekeepers

    Objectives (i)

    } drivers

  • Appreciate the size of the cancer problem and the challenge facing

    patients, clinicians and researchers

    Incidence

    Mortality

    Recognise common cancer terminology

    Apreciate some causes of cancer epidemiology

    Cancer is a genetic disease

    Oncogenes

    Tumour suppressor genes

    DNA repair genes/mechanisms gatekeepers

    An understanding of the processes of tumourigenesis and the

    hallmarks of cancer

    Objectives (i)

    } drivers

  • CANCER; TUMOUR; NEOPLASM

    Benign: normal chromosomes

    differentiated

    rare division/slow growing

    encapsulated

    do not metastasise

    Malignant: abnormal chromosomes

    poorly differentiated

    may be frequent division

    capable of invasion and

    metastasis

    All multicellular organisms can be afflicted by cancer:

    Apparent rising incidence - expanding knowledge - earlier diagnosis

    - increased lifespan

  • An estimated 12.7 million new cancer cases were diagnosed worldwide in 2008.

    Lung, female breast, colorectal and stomach cancers were the most commonly diagnosed

    cancers, accounting for >40% of all cases.

    Worldwide, an estimated 7.6 million deaths from cancer occurred in 2008.

    Lung, stomach, liver, colorectal and female breast cancers were the most common causes, accounting

    for >50% of all cancer deaths.

    Cancer: Global Statistics (2008)

  • Cancer: The U.K. Statistics

    Cancer will affect one in three of the UK population, and be responsible for one in four deaths (>324K new cases in UK in 2010)

    There are more than 200 different types of cancer, but four of them lung, breast, colorectal and prostate account for over half of new cases

    65% of cases occur in those over 65

    In children, leukaemia is the most common cancer

  • Cancer Incidence statistics 2005

  • Cancer Mortality statistics 2007

    Female Male

  • 5 year Cancer Survival 2001 (England and Wales)

    Breast 79%

    Prostate 61%

    Lung 6%

    CRC

  • 21 Common Cancers: 2005 - 2009 and Followed up to 2010 Five-Year Relative Survival, Adults Aged 1599, England

    Breast >80%

    Prostate >80%

    Lung

  • Cancer Classification

    Carcinomas: the most common types of cancer, arise from cells of embryonic endoderm or ectoderm, cover external and internal body surfaces, e.g. epithelia of lung, breast, colon

    Sarcomas: arise from cells of embryonic mesoderm, the supporting tissues of the body such as bone, cartilage, fat, connective tissue and muscle

    Lymphomas: cancers that originate in the lymph nodes and tissues of the bodys immune system

    Leukaemias: cancers of the immature wbcs that grow in the bone marrow and accumulate in large numbers in the bloodsteam

  • Cancer: General Considerations

    Cancer is a disease of abnormal differentiation:

    consider leukaemia, neuroblastoma

    Cancer arises from loss of normal growth control

    Cancer is a disease of genetic origin

    A tumour arises from one ancestral genetically

    aberrant cell

    Tumourigenesis is a multi-stage process

  • Cancer: General Considerations

    Cancer is a disease of abnormal differentiation:

    consider leukaemia, neuroblastoma

    Cancer arises from loss of normal growth control

    Cancer is a disease of genetic origin

    A tumour arises from one ancestral genetically

    aberrant cell

    Tumourigenesis is a multi-stage process

  • Apoptosis

    Proliferation

    Homeostasis perturbed

    Homeostasis

    Proliferation Apoptosis (Cell division) (Cell death)

    cancer

  • Cancer: General Considerations

    Cancer is a disease of abnormal differentiation:

    consider leukaemia, neuroblastoma

    Cancer arises from loss of normal growth control

    Cancer is a disease of genetic origin

    A tumour arises from one ancestral genetically

    aberrant cell

    Tumourigenesis is a multi-stage process

  • Cellular

    metabolism

    UV

    light

    Ionising

    radiation Chemical

    exposure Replication

    errors

    DNA damage: Single strand break

    Double strand break

    Chemical crosslink

    DNA-protein crosslink

    DNA-DNA crosslink

    Base oxidation, alkylation

    Abasic site

    Pyrimidine dimer

    >200 000 events / cell / day

    Cell cycle

    checkpoint

    activation

    Apoptosis

    DNA Repair:

    Direct repair

    Base Excision Repair (BER)

    Nucleotide Excision Repair (NER)

    Mismatch Repair (MMR)

    Homologous Recombination

    Non Homologous End Joining

    DNA Repair

    Genetic origins of Cancer

  • Cancer: General Considerations

    Cancer is a disease of abnormal differentiation:

    consider leukaemia, neuroblastoma

    Cancer arises from loss of normal growth control

    Cancer is a disease of genetic origin

    A tumour arises from one ancestral genetically

    aberrant cell

    Tumourigenesis is a multi-stage process

  • Tumourigenesis is a multistep process:

    May take decades

    Age-dependent cancer incidence e.g. prostate cancer

    Initiation Single genetic event

    Promotion Clonal expansion

    Progression Increased genetic / chromosomal instability,

    accumulation of mutations, increasingly aggressive

    phenotype

    Metastasis

    *

    * *

  • Causes of Cancer

    Tobacco Body weight Physical activity Diet Hormones (reproductive factors - late age at first

    pregnancy a factor in breast cancer)

    Sunlight (UV radiation) Occupational carcinogens (asbestos, benzene,

    pesticides)

    Infectious agents - Viruses (papillomavirus (HPV) in cervical cancer)

    - Bacteria (H. Pylori gastric ulcers/cancer

    Medical treatment (radio- and chemotherapy)

    Pollution (diesel exhaust, xenoestrogens) Heritable cancer predisposition syndromes

  • Smoking- and lung cancer incidence in the

    20th Century

    Cigarette smoke is a toxic

    cocktail of > 4000 chemicals.

    >43 known carcinogens are

    inhaled.

  • Smoking- and lung cancer incidence in the

    20th Century

    Cigarette smoke is a toxic

    cocktail of > 4000 chemicals.

    >43 known carcinogens are

    inhaled.

    HO

    OH

    O

    Benzopyrene[a]pyrene from cigarette

    smoke condensate damages DNA of

    lungs contributing to pathogenesis of lung

    cancer

  • Smoking- damages many parts of the body

  • Causes of Cancer

    Tobacco Body weight Physical activity Diet Hormones (reproductive factors - late age at first

    pregnancy a factor in breast cancer)

    Sunlight (UV radiation) Occupational carcinogens (asbestos, benzene,

    pesticides)

    Infectious agents - Viruses (papillomavirus (HPV) in cervical cancer)

    - Bacteria (H. Pylori gastric ulcers/cancer

    Medical treatment (radio- and chemotherapy)

    Pollution (diesel exhaust, xenoestrogens) Heritable cancer predisposition syndromes

  • Infectious causes of cancer

    Oncogenic human viruses:

    Hepatitis C RNA virus

    Causes hepatocellular carcinoma through

    cycles of inflammation, repair and regeneration.

    Oncogenic bacteria:

    Heliobacter pylori, a spiral flagellated

    Gram negative bacterium which underlies

    pathogenesis of gastric carcinoma

    causes gastritis, peptic ulceration and cancer.

    Half the world`s population infected

    Barry Marshall

    http://discovermagazine.com/2010/mar/07-dr-drank-broth-gave-

    ulcer-solved-medical-mystery

  • 1. Self-sufficiency in growth signals

    2. Insensitivity to growth-inhibitory signals

    3. Evasion of programmed cell death

    (apoptosis)

    4. Limitless replicative potential

    5. Sustained angiogenesis

    6. Tissue invasion and metastasis

    7. Reprogramming energy metabolism

    8. Evasion of immune destruction

    The Hallmarks of Cancer (Hanahan and Weinberg, Cell, 2011, 144, 646-674)

    Cancer is complex but can be understood in terms of

    underlying Principles of Tumourigenesis.

  • Apoptosis

    Proliferation

    Homeostasis perturbed

    Homeostasis

    Proliferation Apoptosis (Cell division) (Cell death)

    cancer

  • Homeostasis

    Proliferation Apoptosis

    Proto-Oncogenes

    Tumour suppressor genes

    C-abl p53

    Cancer cells acquire mutations in genes which encode

    proteins whose roles in regulatory circuits control

    normal cell proliferation and homeostasis

  • Homeostasis

    perturbed

    Proliferation

    Carcinogenic Oncogenes

    Apoptosis

    Tumour suppressor genes

    bcr-abl

    p53

    X

    Cancer

  • Proto-oncogenes and Signalling Systems

    Proto-oncogenes encode proteins that relay growth-stimulatory signals from outside the cell to the nucleus

    Growth factors bind to specific receptors on the cell membrane

    Receptor binding activates proliferative signal transduction cascades within the cytoplasm

    The succession of relay proteins results in activation of transcription factors which drives transcription of genes producing proteins which usher the cell through its growth cycle

  • Oncogenes and Signalling Systems A Some oncogenes force cells to

    overproduce growth factors e.g. sarcomas and gliomas release excessive platelet-derived growth factor

    B Some oncogenes produce aberrant or elevated receptors which release proliferative signals even in the absence of growth factor, e.g. Erb-B2 (Her2) receptors in breast cancer cells

    C Some oncogenes perturb signalling cascades in the cytoplasm, e.g. mutant Ras protein found in carcinomas of the colon, pancreas and lung

    D Some oncogenes alter the activity of nuclear transcription factors, e.g. c-myc in the absence of growth factors in leukaemias, breast, stomach and lung cancers

    Ras

    Her2

    Her2

    cytoplasm

    nucleus

    c-myc

  • The Hallmarks of Cancer (1)

    Self-Sufficiency in Growth Signals

    Proto oncogenes Oncogenes

    mutated

    inappropriately expressed

    constitutively activated

    Cancer

  • q 34

    q 11 bcr

    c-abl proto-oncogene

    145 kDA

    Chr 9 22 9q+ Ph1

    t (9;22)

    Translocation mutation

    bcr-abl 210 kDa

    Carcinogenic oncogene

    - tyrosine kinase activity

    constitutively active

    9q+ Ph1

    Pathogenesis of Chronic Myeloid Leukaemia (CML)

  • Point Mutations:

    If a point mutation occurs, - a single base is altered

    - an altered codon

    - different amino acid

    - protein function

    Point mutations in codons 12, 13 or 61 lead to constitutive activation of growth stimulatory signals and tumour formation:

    Codon 61: CAA AAA; glutamine - lysine

    Ras Most common carcinogenic oncogene in human

    cancer (25%): 90% pancreas, 50% colon; 50% thyroid.

  • The Hallmarks of Cancer (2):

    Insensitivity to growth-inhibitory signals

    Cancer cells evade or ignore braking signals issued by normal cells

    Tumour suppressor genes (P53, PTEN) are mutated, thereby inactivated

    In a point mutation: - a single base is altered

    - an altered codon

    - different amino acid

    - protein function

    In P53 protein: G A; codon 273 results in Arginine - Histidine and inactivation of function.

  • The Hallmarks of Cancer (2):

    Insensitivity to growth-inhibitory signals

    P53 protein (the guardian of the genome) can halt cell cycle progression in response to DNA

    damage, promoting DNA REPAIR or inducing

    APOPTOSIS (programmed cell death)

    Mutant p53 is found in over half of all human tumours

  • You have access to different types of cancer either

    via the menu on the left or by clicking on a specific

    cancer name on the map below.

  • Cell Cycle

    G1 Production of RNA proteins

    and enzymes needed for

    DNA synthesis

    S DNA synthesis

    G2 preparation for mitosis,

    specialised proteins are

    synthsised

    M prophase, metaphase,

    anaphase, telophase

    Division to form 2 daughter

    cells.

    P53

    P53

    p53

    Division of cells with carcinogenic DNA lesions

    X Cell cycle arrest DNA Repair MMR,

    BER,

    NER,

    HR

  • The Hallmarks of Cancer (3):

    Evasion of programmed cell death (apoptosis)

    Tumour growth is determined not only by the rate of cell proliferation but also by the rate of cell attrition, Programmed Cell Death: APOPTOSIS

    P53 protein helps to trigger cell suicide; its inactivation by many tumour cells reduces the likelihood of elimination of damaged cells

    Cancer cells also synthesise excessive amounts of survival proteins (eg Bcl-2), which inhibit apoptosis

  • Cellular

    metabolism

    UV

    light

    Ionising

    radiation Chemical

    exposure Replication

    errors

    Genetic origins of Cancer

    P53

    Apoptosis

    X

    Cells with damaged DNA, harbouring

    carcinogenic lesions survive and proliferate

  • Cellular

    metabolism

    UV

    light

    Ionising

    radiation Chemical

    exposure Replication

    errors

    Genetic origins of Cancer

    X

    Cells with damaged DNA, harbouring

    carcinogenic lesions survive and proliferate

    BCL-2

    MCL-1

    SURVIVAL

  • :

    Proto-oncogenes

    Tumour supressor genes

    Genes encoding proteins involved in DNA repair:

    Nucleotide Excision Repair (NER): XP

    Homologous Recombination: BRCA1/2

    DNA Damage leading to Genetic Mutations.

    Tumour initiation

    Cellular

    metabolism

    UV

    light

    Ionising

    radiation Chemical

    exposure Replication

    errors

  • Li-Fraumeni syndrome: Heritable Cancer Predispositions conferred by germline

    mutation in P53.

    Germline mutation in P53

    tumour suppressor gene 17p13

    Predisposes sufferers to

    wide variety of cancer types

    Young age at onset of

    malignancies

    Multiple primary sites during

    lifetime:

    Breast, brain, leukaemia, soft

    tissue / bone sarcoma.

    BBC Radio 4 Inside the Ethics Committee Series 9 episode 3; 22. 08. 2013.

  • So far

    Cancer is a genetic disease

    Oncogenes

    Tumour suppressor genes

    Genes encoding DNA repair proteins

    Hallmarks of cancer

    Self sufficient growth

    Overcome growth inhibitory signals

    Evasion of apoptosis

  • Hallmarks of cancer

    Self sufficient growth

    Overcome growth inhibitory signals

    Evasion of apoptosis

    Limitless replicative potential

    Sustained angiogenesis

    Invasion

    Metastasis

  • DNA segments at the ends of chromosomes,

    known as telomeres (TTAGGG in humans)

    protect chromosomal integrity, tally the

    number of replicative generations and initiate

    senescence and crisis.

    The Hallmarks of Cancer (4):

    Infinite replicative potential

    Thus, the number of divisions a cell can

    undergo is finite

    The telomerase enzyme complex restores

    telomeres to the ends of chromosomes,

    enabling endless replication

    Telomerase is active in >85% cancers but

    inactive in most normal cells

  • The Hallmarks of Cancer (4):

    Infinite replicative potential

  • The Hallmarks of Cancer (5):

    Sustained angiogenesis

    Proliferation of new capillaries is known as angiogenesis (or neovascularisation), it is typically short-lived (one or two weeks).

    Tumour cells however can switch on angiogenesis. Angiogenesis is crucial for:

    Primary tumour growth (promotion of a small cluster of mutated cells to a large malignant growth)

    Metastasis.

  • Blood vessel

    Oxygenated area

    Hypoxic tumour mass

    Necrotic region

    Why is angiogenesis important for tumours?

  • In situ carcinoma: undetectable microscopic mass of tumour cells

    Tumour cells secrete molecules (e.g. vascular endothelial growth

    factor, VEGF)

    Signalling activates genes/ protein synthesis in normal host tissue

    encouraging growth of new blood

    vessels to the tumour

  • In normal tissues, cells adhere to cells and to extracellular matrix (ECM): growth is anchorage dependent.

    In cancer cells, cell adhesion molecules: E-cadherin are compromised or absent. Growth becomes anchorage independent.

    Cancer cells release matrix metalloproteinase enzymes which dissolve basement membranes allowing invasion and escape.

    Invasion of vascular beds of distant organs allows secondary tumours, metastases to establish.

    The Hallmarks of Cancer (6):

    Tissue Invasion and metastasis

  • Cancers are capable of spreading through the body by:

    Invasion: Penetration of cancer cells into neighbouring tissues:

    Mammary carcinoma invading the surrounding breast tissue

    The Hallmarks of Cancer (6):

    Tissue Invasion and metastasis

  • (6) Tissue Invasion and metastasis

    Secondary tumours are more aggressive and resistant to therapy.

    Metastases account for ~ 90% cancer deaths.

    Metastasis: Systemic circulation and invasion of distant tissues:

    lung,

    liver,

    bone.

  • Cancer is a genetic disease

    Oncogenes

    Tumour suppressor genes

    Genes encoding DNA repair proteins

    Hallmarks of cancer

    Self sufficient growth

    Overcome growth inhibitory signals

    Evasion of apoptosis

    Infinite replicative ability

    Sustained angiogenesis

    Invasion and metastasis

    Cell 2011 Mar 4;144(5):646-74. D Hanahan and RA Weinberg Hallmarks of cancer: the next generation.

  • Current Treatment Options

    Surgery (localised tumour)

    Radiotherapy (localised tumour)

    Chemotherapy (disseminated tumour)

  • Cancer Chemotherapy

    Chemotherapy uses chemical agents, which may be:

    natural products, natural product-derived (semi- synthetic), natural product-inspired,

    synthetic small molecule or antibody anticancer drugs to destroy or control the growth of cancer cells.

    Because chemotherapy treats the whole body, it can

    treat cancer cells that have already escaped the

    primary tumour.

  • Objectives

    Understand the mechanisms of action of major clinically used cytotoxic anticancer drugs.

    A knowledge of mechanisms of acquired / inherent resistance to chemotherapeutic agents, and

    appreciate rationale underpinning combination

    therapy.

    Appreciate the side effects associated with chemotherapeutic agents and their cause.

  • Cell Cycle

    G1 Production of nucleotide

    bases, RNA proteins and

    enzymes needed for DNA

    synthesis

    S DNA synthesis

    G2 preparation for mitosis,

    specialised proteins are

    synthesised

    M prophase, metaphase,

    anaphase, telophase

    Division to form 2 daughter

    cells.

    G0 Dormant phase

    G0

  • Cytotoxic chemotherapy interferes primarily with DNA synthesis and mitosis (the (G1) S and M phases of the

    cell cycle) to destroy cancer cells.

    Chemotherapeutic drugs do not distinguish between normal cells and cancer cells. Adverse reactions to

    chemotherapy are a consequence of cytotoxicity to normal cells.

    However, cytotoxic chemotherapeutic agents do show selectivity for cancer cells over normal cells:

    Normal cells are able to repair DNA; normal tissues

    recover. Injury caused by chemotherapy is rarely

    permanent.

  • Mustard Gas was first used in September 1917 during WWI. It was one of the most

    lethal of all the poisonous chemicals used in warfare.

    British soldiers blinded by mustard gas

    Cytotoxic Chemotherapy: History

  • During WWII, it was found that soldiers who were

    exposed to mustard gas, sulfur mustard suffered

    from lower white blood cell counts.

    This discovery led to the use of nitrogen mustard, a

    similar but less toxic chemical agent, to cure patients

    with high white blood cells counts (lymphoid

    leukaemia) and lymphomas.

    Cytotoxic Chemotherapy: History

    S

    CH2--CH2--Cl

    CH2--CH2--Cl

  • During WWII, it was found that soldiers who were

    exposed to mustard gas, sulfur mustard suffered

    from lower white blood cell counts.

    This discovery led to the use of nitrogen mustard, a

    similar but less toxic chemical agent, to cure patients

    with high white blood cells counts (lymphoid

    leukaemia) and lymphomas.

    Cytotoxic Chemotherapy: History

    S

    CH2--CH2--Cl

    CH2--CH2--Cl

    N

    CH2--CH2--Cl

    CH2--CH2--Cl

    H3C

    In 1942, Nitrogen Mustard chlormethine, became the first antineoplastic chemotherapeutic agent used medicinally.

    This agent forms a covalent bond with DNA,

    Alkylating specific sites on purine (A, G) bases of DNA

    Crosslinking DNA

    Causing cell death

    This ALKYLATING AGENT is the pioneer of Antineoplastic Chemotherapy

  • 4 (nucleo)bases: A G C T

    Base + deoxyribose sugar = nucleoside

    Base + deoxyribose sugar + phosphate =

    nucleotide

    Nucleotide polymers = nucleic acid (DNA)

    Deoxyribonucleic acid (DNA)

  • Nuceotide polymers:

    DNA double helix

  • CH3

    N7 methyl guanine

    Nuceotide polymers:

    DNA double helix Pyrimidines

    Purines

  • Alkylating agents

    The oldest class of anticancer drugs

    Major cornerstone of treatment for leukaemias, lymphomas and solid

    tumours.

    Other nitrogen mustard analogues:

    Chlorambucil

    Melphalan,

    Ifosfamide

    Cyclophosphamide:

    One of most widely used cytotoxic agents in combination or sequentially with other antineoplastic drugs. Used to treat cancers of:

    Brain, breast, bladder, cervix, endometrium, lung, testis, ovary, Burkitt`s, and other

    non-Hodgkin`s lymphoma, multiple myeloma, gestational trophoblastic tumours,

    childhood malignancies neuroblastoma, retinoblastoma, Wilms`, Ewing`s, leukaemias: CLL, ALL, CML, AML.

    NCH2--CH2--Cl

    CH2--CH2--ClHO2C(H2C)3

    NH

    P

    O

    NCH2--CH2--Cl

    CH2--CH2--Cl

    O

  • Cyclophospahmide - Mechanism of action:

    Cyclophosphamide is a prodrug requiring biotransformation by

    cytochrome P450 (CYP 3A4 in the liver) to exert toxicity.

    NH

    P

    O

    NCH2--CH2--Cl

    CH2--CH2--Cl

    ONH

    P

    O

    NCH2--CH2--Cl

    CH2--CH2--Cl

    O

    HO

    CYP 3A4

    4-OH-CPA

    NCH2--CH2--Cl

    CH2--CH2--ClP

    O

    H2N

    O-

    H

    O

    +

    Phosphoramide mustard + acrolein

    active alkylating species unwanted by-product

    Cross-links RNA and DNA haemorrhagic cystitis

  • Alkylating agents 2: Nitrosourea derivatives

    The anticancer activity of this class of compounds was discovered in 1959 at the NCI

    Activity against solid and non-solid tumours

    Lipophilic allowing access across BBB

    Chloroethylate DNA

    Therapeutic efficacy limited by development of resistance, involving multiple repair pathways

    ClNNO

    NH

    Cl

    O

  • Alkylating agents 2: Nitrosourea derivatives

    The anticancer activity of this class of compounds was discovered in 1959 at the NCI

    Activity against solid and non-solid tumours

    Lipophilic allowing access across BBB

    Chloroethylate DNA

    Therapeutic efficacy limited by development of resistance, involving multiple repair pathways

    ClNNO

    NH

    Cl

    OCarmustine

    Used alone or adjuvant treatment of

    brain, colon, lung cancers, Hodgkin`s,

    non-Hodgkin`s lymphoma, melanoma,

    multiple myeloma, mycosis fungoides.

  • Alkylating agents 2: Nitrosourea derivatives

    The anticancer activity of this class of compounds was discovered in 1959 at the NCI

    Activity against solid and non-solid tumours

    Lipophilic allowing access across BBB

    Chloroethylate DNA

    Therapeutic efficacy limited by development of resistance, involving multiple repair pathways

    Carmustine

    Used alone or adjuvant treatment of

    brain, colon, lung cancers, Hodgkin`s,

    non-Hodgkin`s lymphoma, melanoma,

    multiple myeloma, mycosis fungoides.

    Antineoplastic and toxic effects caused by active metabolites. Chloroethyl carbonium

    ion leads to formation of DNA cross links during all phases of cell cycle, causing cell

    cycle arrest and apoptosis.

  • N N

    NN

    N

    CH3

    H2NOC

    O

    Alkylating agent

    Treatment of high grade Glioma recurrent anaplastic astrocytoma

    and gliobastoma multiforme

    Malignant melanoma and brain

    metastases from solid tumours

    Alkylating agents 3: Triazenes

    NH

    NN

    N

    H2NOC

    N

    CH3

    CH3

    Alkylating agent

    Treatment of metastatic

    malignant melanoma (response

    rate 15-25%)

    Hodgkin`s lymphoma (in combination with doxorubicin,

    bleomycin and vinblastine)

    Dacarbazine Temozolomide

  • Malignant Melanoma

  • Ed Newlands

  • 11C (Me)-Temozolomide in positron emission tomography (PET) after oral administration

    Saleem et al, Cancer Res., 2003, 63, 2409-2415

  • 11C methylation of DNA

    N N

    N

    N

    O

    N

    H2NOC

    Me14C 11C

    11C

    15N

    Isotopic labelling of temozolomide

    Expired 11CO2

    Expired N2

    NH

    N

    H2NOC

    14C

    NH2

    Excretion

    Protonation

  • N N

    NN

    N

    CH3

    H2NOC

    O

    Major Mechanism of Resistance inherent or acquired:

    Repair of methylated DNA O6methyl guanine by methyl guanine methyl transferase MGMT.

    Tumours whose cells express MGMT are resistant to

    Temozolomide

  • Alkylating Agents

    Transfer an alkyl group onto DNA

    CH3 methyl

    CH3CH2Cl chloroethyl

    DNA damage :

    in S phase (block DNA synthesis), G2

    arrest

    DNA single strand breaks- double

    strand breaks.

    Cross link the 2 strands.

  • Antimetabolites

    Bastion of Cancer Chemotherapy

    Chemicals structurally similar to either FOLATE or NUCLEOBASES

    DNA building blocks

    FOLIC ACID growth factor, provides

    carbon atoms for nucleotide precursors

    in synthesis of RNA and DNA

    FOLATE ANTAGONISTS inhibit one

    (or more) folate-dependent enzymes

  • Purine antagonists inhibit

    production of A and G in DNA and

    RNA

    Guanine

    Adenine

    Cytosine

    Thymine

    Uracil

    Pyrimidine antagonists block

    synthesis of C and T in DNA; C

    and U in RNA

    Thus, the antimetabolites

    BLOCK DNA SYNTHESIS

  • Methotrexate

    Most commonly used folate antagonist.

    Enters cell via folate receptors

    Binds DHFR

    Inhibits synthesis of FH4

    Cell cannot create new purine and thymidine nucleotides

    DNA and RNA synthesis blocked

  • Methotrexate

    Administered alone or in combination with other chemotherapeutic agents to treat:

    leukaemia, lymphoma

    gestational choriocarcinoma

    solid tumours: breast, head and neck, lung, bladder,

    oesophagus

    Methotrexate can be given by many routes: po, iv, sc, im, intra-arterial, intrathecal.

    It is the only cytotoxic drug for which there is a role for routine PK monitoring:

    leucovorin / folinic acid rescue bypasses inhibited DHFR,

    replenishing intracellular reduced folate pools.

    Other clinical folate antagonists: Permetrexed, Ralitrexed, Nolatrexed

  • Pyrimidine Analogues

    5-FLUOROURACIL and CAPECITABINE decrease the biosynthesis of

    pyrimidine nucleotides by inhibiting thymidylate synthase, the enzyme

    that catalyses the rate limiting step in DNA synthesis.

    CAPECITABINE is converted to 5-FLUOROURACIL in cancer cells

    5-FU cannot be methylated by thymidylate synthase, causing sustained

    inhibition of the enzyme and decreased production of Thymine.

    5-FU is incorporated into DNA (via dUMP or dTMP) or RNA (via UTP),

    leading to cytotoxicity via DNA strand breakage and decrease in protein

    synthesis

    5-FU

  • Pyrimidine Analogues

    5-FLUOROURACIL and CAPECITABINE decrease the biosynthesis of

    pyrimidine nucleotides by inhibiting thymidylate synthase, the enzyme

    that catalyses the rate limiting step in DNA synthesis.

    Clinical Applications:

    In a number of iv combination chemotherapies for treatment of bowel,

    breast, gastric, oesophageal, pancreatic, head and neck, anal and

    ovarian cancers.

    Oral analogues are also used, and topical application for superficial basal

    cell carcinoma.

    5-FU

    Additional pyrimidine antagonists: Gemcitabine, Tegafur, Sapacitabine

  • Purine Antagonists

    In clinical use since 1953

    Mercaptopurine

    Adenine analogue

    Treatment of ALL, AML, Hodgkin`s lymphoma in children,

    lymphoblastic lymphoma.

    Dose-related toxicity bone marrow suppresssion

    Thioguanine

    Synthetic guanine analogue inhibition of purine synthesis

  • Antimetabolites

    Inhibit production of DNA building blocks

    Antifolates

    Purine antagonists

    Pyrimidine antagonists

    Act in G1 cell cycle phase

    (Prevent DNA synthesis)

  • Antimicrotubule agents

    The Company of Biologists Ltd 2005

    Paralyse tubulin (which forms

    microtubules) or microtubules

    Critical for cell division

    Block cell cycle during Mitosis

    Microtubules allow segregation of chromosomes during mitosis

  • Antimicrotubule agents 1

    Paclitaxel (Taxol) Isolated from Pacific Yew:

    Taxus brevifolia

    Problem: limited supply and extraction

    Semi-synthetic Docetaxel (Taxotere) from the more abundant taxane precursor

    found in needles of European Yew:

    Taxus baccata

    Taxanes

    Microtubule poisons

    - stabilise microtubules

    - inhibit depolymerisation

    cause G2/M cell cycle arrest

    Treatment of Breast, Ovarian, Lung carcinomas

  • Antimicrotubule agents 2

    Vinca Alkaloids

    Isolated from Madagascan

    periwinkle Catharanthas roseus.

    > 70 alkaloids identified in sap

    from C roseus used historically in

    folk (traditional) medicine.

    Vincristine (Oncovin) Vinblastine (Velbe)

    Used in treatment of leukaemia, lymphoma,

    melanoma, soft tissue sarcoma,

    neuroblastoma, breast and lung cancers.

    Mechanism of action:

    Bind -tubulin

    Inhibit tubulin polymerisation

    Destabilise (depolymerise)

    microtubules:

    Destruction of mitotic spindle, leaving

    cells stranded in mitosis.

  • Antimicrotubule agents

    The Company of Biologists Ltd 2005

    Tubulin Microtubules Polymerisation

    depolymerisation

    Vinca alkaloids

    Taxanes

    Preventing cells from completing mitosis

  • control

    taxol

    vincristine

    Antimicrotubule agents

    Prevent successful mitoses

  • Cytotoxic chemotherapy So far.

    Alkylating agents Nitrogen mustard

    Cyclophosphamide

    Temozolomide

  • Cytotoxic chemotherapy So far.

    Alkylating agents Nitrogen mustard

    Cyclophosphamide

    Temozolomide

    Antimetabolites Methotrexate folate antagonist 5-Fluorouracil pyrimidine antagonist Thioguanine purine antagonist

  • Cytotoxic chemotherapy So far.

    Alkylating agents Nitrogen mustard

    Cyclophosphamide

    Temozolomide

    Antimetabolites Methotrexate folate antagonist 5-Fluorouracil pyrimidine antagonist Thioguanine purine antagonist

    Spindle poisons Taxanes stabilise microtubules Vinca alkaloids inhibit tubulin polymerisation

    Anticancer Therapeutics: Ed Sotiris Missailidis, Wiley-Blackwell, 2008.

  • Platinum drugs

    Pt

    H3N

    Cl

    Cl

    H3N

    Cisplatin

    Discovery 1965

    Clinical application 1970s

    Intravenous short term infusion in saline

    Treatment of solid/epithelial malignancies:

    First line and in combination: testicular, ovarian, cervix uterus, lung, head and neck, oesphagus, stomach, colon,

    bladder.

    Second line: advanced breast, pancreas, liver, kidney, prostate,

    peritoneal and pleural mesotheliomas.

  • Cisplatin: Mechanism of Action:

    Cisplatin crosslinks DNA causing interstrand and intrastrand GG,

    AG Pt adducts.

    Replication inhibition,

    Transcription inhibition,

    Cell cycle arrest,

    DNA repair mechanisms

    triggered - failure,

    Cell Death

  • Carboplatin

    Adverse Cisplatin reactions

    Renal, GastroIntestinal Toxicity, Peripheral Neuropathy.

    Markedly less toxic to kidneys and nervous system causing less nausea and vomiting.

  • Platinum agents

    Cross link DNA

    Damage triggers cell cycle arrest and DNA repair mechanisms

    Which fail (initially)

    Activating apoptosis

    S cell cycle phase

  • Antitumour Antibiotics

    Substances produced by micro-organisms (or plants, or

    marine invertebrates) that exert anticancer activity by

    DNA interaction.

    Multiple modes of action, may be subject to alternative

    classification - alkylating agents

    - DNA cleavage agents

    - non covalent binding agents

    - topoisomerase inhibitors

    Actinomycin D

    Mitomycin C

    Bleomycin

    Doxorubicin - anthracycline

    Etoposide - podophyllotoxin

    Irinotecan - camptothecin

  • Bleomycin

    Linear glycosylated peptide antibiotic

    Isolated from Streptomyces verticellus in 1960s

    There are > 200 members of bleomycin family,

    blenoxane = bleomycin A2 + bleomycin B2

    Treatment of: Hodgkin`s, non-Hodgkin`s lymphomas, squamous

    cell carcinoma, testicular carcinoma, malignant

    pleural effusions, Kaposi`s sarcoma.

    Mechanism of Action:

    DNA damage generation of DNA Double Strand Breaks (DSBs) and single strand breaks (SSBs).

    RNA cleavage caused by bleomycin-induced oxidative damage.

    Nucleic acid cleavage dependent on presence of Fe2+ and molecular

    oxygen.

  • Bleomycin

    Linear glycosylated peptide antibiotic

    Isolated from Streptomyces verticellus in 1960s

    There are > 200 members of bleomycin family,

    blenoxane = bleomycin A2 + bleomycin B2

    Treatment of: Hodgkin`s, non-Hodgkin`s lymphomas, squamous

    cell carcinoma, testicular carcinoma, malignant

    pleural effusions, Kaposi`s sarcoma.

    Serious Adverse Reaction:

    Cumulative pulmonary toxicity Lung fibrosis

    Mortality in 1-2% bleomycin treated patients.

    Manageable adverse reactions:

    nausea, vomiting, appetite loss, alopoecia, allergic reactions, skin damage

  • Doxorubicin

    Anthracycline antibiotic tetracyclic chromophore with

    anthraquinone motif.

    Antibacterial,

    Immunosuppressive,

    Antiparasitic and wide spectrum

    Antitumour activity.

    First isolated in 1960s from

    cultures of Streptomyces peucetius.

    Large scale prep developed in 1970s involve semisynthetic

    process starting from daunoubicin

    Doxorubicin and Daunorubicin, used clinically for four

    decades, are amongst most efficient antitumour drugs.

  • Doxorubicin

    Frequent Indications include:

    Breast, ovarian, transitional cell bladder cancer, bronchogenic

    lung cancer, thyroid and gastric cancer, soft tissue and

    osteogenic sarcomas, neuroblastoma, Wilms` tumour,

    malignant lymphoma (Hodgkin`s and non Hodgkin`s, acute

    myeoloblastic leukaemia, acute lymphoblastic leukaemia and

    Kaposi`s sarcoma

    Clinical Limitations:

    Development of Acquired Resistance

    Cardiotoxicity. Dilative cardiomyopathy and congestive heart

    failure are cumulative adverse effects that appear (usually) after 1 year.

    To prevent these severe reactions, a maximum cumulative dose of 600

    mg/m2 has been established.

    Less severe side effects include nausea, vomiting, diarrhoea, loss of

    appetite, hair loss and skin damage.

  • Mechanism of Action of Doxorubicin

    Doxorubicin is a Topoisomerase II poison/inhibitor

    DNA exists in cells as a supercoiled double helix which must unwind during

    transcription and replication.

    Topoisomerse enzymes catalyse changes in DNA topology.

    Doxorubicin inhibits Topoisomerase II-catalysed DNA relaxation

    Doxorubicin binds strongly to duplex DNA by intercalation selectively at C-G sequences.

    generation of DNA DSBs inhibiton of religation of cleaved duplex

    Halted DNA synthesis

    Cell Death

    Doxorubicin leads to generation of Reactive Oxygen Species (ROS) which damage DNA

    Formation of DNA adducts and crosslinks

    Inhibition of DNA and RNA synthesis

  • Podophyllotoxin isolated from Podophyllum

    peltatum Mayapple 1880

    Potent antitumour activity with severe side effects

    no clinical use

    Podophyllotoxin - Etoposide

    Research led to

    synthesis of

    Podophyllotoxin

    analogues Etposide,

    Teniposide

  • Mechanism of Action

    Podophyllotoxin: Binds tubulin and inhibits mitotic spindle assembly

    Etoposide and Teniposide poison the Topoisomerase II-DNA complex.

    (forming a ternary complex) and prevent re-ligation of DNA

    DNA Double Strand Breaks

    Cell Death

    Etoposide Clinical Applications

    Main indications are for testicular and lung cancers

    Non Hodgkin`s, Hodgkin`s lymphoma, AML, ALL, CML, mycosis

    fungoides, Wilms`, neuroblastoma, Kaposi`s, gestational trophoblastic

    tumours, ovarian germ cell tumours,brain and refractory breast cancer.

    Dose related and limiting myelosuppression. Bone marrow recovery

    usually complete (3 weeks).

  • Camptothecin

    Pentacyclic antitumour antibiotic isolated

    1966 from alkaloid extracts of Camptotheca acuminata.

    Remarkable activity against murine

    leukaemia models (NCI)

    Mechanism of Action

    Camptothecin binds and stabilises DNA-Topisomerase I complex,

    inhibiting religation of cleaved DNA strands, blocking DNA

    synthesis.

    In the 1990s, water soluble camptothecin analogues were

    synthesised, demonstrating Topo I inhibition and potent antitumour

    activity: Topotecan and Irinotecan have reached the clinic.

  • Irinotecan

    Approved in Japan in 1994

    lung,

    cervical

    ovarian cancers

    Subsequently in Europe and US for

    treatment of Colorectal cancers.

    Side effects: diarrhoea, neutropaenia

    Novel camptothecin analogues under clinical evaluation including

    liposome-encapsulated camptothecins.

  • So far

    DNA alkylators Platinum agents

    Antimetabolites Antitumour antibiotics

    Tubulin inhibitors Topisomerase inhibitors

  • Cytotoxic Chemotherapy targets rapidly dividing cells

    DNA alkylators Platinum agents

    Antimetabolites Antitumour antibiotics

    Tubulin inhibitors Topisomerase inhibitors

  • Cytotoxic Chemotherapy targets rapidly dividing cells

    Dose limiting toxicity: GI, haematological, renal, cutaneous toxicities:

    Cells which divide rapidly under normal circumstances vulnerable

    Bone marrow - myelosuppression

    Digestive tract - mucositis

    Hair follicles - alopecia

    Lack of clinical efficacy

    Drug resistance

    inherent, acquired

    .

    Cytotoxic Chemotherapy: Limitations

  • Clinical mechanisms of resistance

    Decreased drug uptake

    down regulation of Cu transporting pump decreased intracellular Cisplatin concentrations and reduced Cisplatin toxicity

    Enhanced drug efflux

    In 1976, the MultiDrug Resistance protein-1 (MDR-1; P-

    glycoprotein (P-gp); ABCB1) was

    described.

    P-gp pumps out of the cell a large number of broadly active

    cytotoxic chemotherapeutic agents.

    Cells overexpressing Pgp possess a

    multidrug resistant phenotype.

  • Enhanced drug efflux

    ATP binding casette (ABC) protein pumps commonly overexpressed in resistant

    tumours, capable of transporting anticancer agents with distinct mechanisms

    of action.

    ABCB1 (P-gp); MRP-1 (multiple drug resistance protein-1; ABCC1); BCRP

    (breast cancer resistance protein, ABCG2); MRP-5.

  • Clinical mechanism of resistance

    Decreased drug uptake

    Enhanced drug efflux (P-gp)

    Altered target expression

    upregulated transcription and overexpression of DHFR in response to Methorexate treatment

    DNA repair MGMT removes and repairs alkylated DNA bases (Temozolomide)

    Failure to activate (pro)drug (cytochrome P-450)

    Metabolic drug inactivation

    Mutations / altered activity of drug target

    (decreased topoisomerase II-etoposide binding affinity;

    Tubulin mutations)

  • Combination Chemotherapy

    Treatment with a number of agents / regimes

    simultaneously / sequentially with DISTINCT

    mechanisms of action:

    To eradicate tumour cells - heterogenous disease

    Prevent emergence of acquired resistance

    Limit toxicities / spare normal cells

  • Acute Lymphoblastic Leukaemia

    80% childhood leukaemia

    85% successful treatment

    Combination treatment comprises:

    Intrathecal MTX - antifolate antimetabolite

    Vincristine vinca alkaloid

    Daunorubicin antitumour antibiotic

    Steroids enhance chemotherapy efficacy, wellbeing, combat tiredness,

    anti-sickness, improve apetite, balance body salts, water

    L-Asparaginase - an enzyme that destroys asparagine external to the

    cell. Normal cells are able to make all the asparagine they need

    internally whereas tumour cells become depleted rapidly and die.

    Radiotherapy

  • Combination Therapy Breast Cancer

    Surgery, Radiotherapy, Chemotherapy.

    Cyclophosphamide alkylating agent

    Epirubicin, Doxorubicin antibiotic, DNA intercalator

    5FU antipyrimidine

    Methotrexate antifolate

    Mitoxantrone topoisomerase II inhibitor

    Taxol antimicrotubule

    Gemcitabine antipyrimidine

    Tamoxifen (Hormone therapy; Herceptin, targeted, biological therapy.

  • Understand the mechanisms of action of major clinically used cytotoxic anticancer drugs.

    Appreciate the side effects associated with chemotherapeutic agents and their cause.

    A knowledge of mechanisms of acquired/inherent resistance to chemotherapeutic agents

    Appreciate rationale underpinning combination therapy.

    Summary Slide

  • Objectives: Target-directed therapy

    Be aware of general treatment strategies.

    Understand the mechanisms of action of major clinically used anticancer drugs.

    Rationale behind target-directed therapy.

    Knowledge of example molecularly targeted therapies and mechanisms of action.

    A knowledge of mechanisms of acquired resistance to targeted agents, and appreciate rationale underpinning combination therapy.

    Appreciate the side effects associated with chemotherapeutic

    agents and their cause.

  • Cytotoxic Chemotherapy

    Targets rapidly dividing cells (DNA)

    DNA alkylators (S; cyclophosphamide; temozolomide)

    Antimetabolites (G1; 5-FU, methotrexate)

    Topisomerase inhibitors (etoposide)

    Antitumour antibiotics (S; bleomycin, doxorubicin)

    Tubulin inhibitors (G2/M; vincristine, taxol)

    Limitations

    Dose limiting side effects:

    GI, haematological, renal, cutaneous toxicities

    Drug resistance

    Lack of clinical efficacy

  • Aim

    To develop targeted agents that are tumour specific with decreased

    toxicity profiles than chemotherapeutic agents.

    Target-directed drug discovery

    Advances in cancer cell and molecular biology have led to identification

    of carcinogenic oncogenes and elucidation of cancer signalling networks

    Patient / tumour

    selection

    development of

    target-directed agents

  • Key Signalling Cascades in tumourigenesis

  • Workman, P. and Kaye, S.B. (2002) Translating basic cancer research into new

    cancer therapeutics. Trends. Mol. Med. 8(4), S1-S9.

    Workman, P. (2005) Genomics and the second golden era of cancer drug development. Molecular

    Biosystems 1(1), 17-26.

    Target-directed drug discovery

    identification of a validated biological target (normally a protein)

    a molecule hunt, typically involving a high-throughput screening

    hit-to-lead development to identify (a) lead compound(s)

    lead development (involving in vivo testing and optimisation of ADMET properties)

    [Absorption, Distribution, Metabolism, Excretion, Toxicity].

    clinical trials with appropriate pharmacodynamic (PD) endpoints

  • Tyrosine kinases (TKs) encoded by proto-oncogenes - are

    important mediators of signalling cascades:

    Key roles in growth, differentiation, metabolism and apoptosis

    TK activity is tightly regulated in normal cells

    Role of TKs in pathophysiology of cancer:

    Mutation(s), overexpression of TKs (carcinogenic oncogenes)

    lead to malignant growth.

    Block constitutive activation of TKs in cancer cells:

    Selective Small Molecule and

    Monoclonal Antibody Inhibitors:

    Tyrosine Kinases - Roles and significance in Cancer.

  • HER2

    } EGFR

    bcr/abl

    Small molecule RTK Antibody

    Gleevec bcr/abl

    Vemurafenib BRAF

    HER2 Herceptin

    Iressa EGFR Erbitux

    Tarceva

    Sunitinib VEGFR Avastin

    Tyrosine Kinase inhibitors examples

    Targeted treatment for:

    CML Melanoma Breast Prostate NSCLC CRC

  • q 34

    q 11 bcr

    c-abl proto-oncogene

    145 kDA

    Chr 9 22 9q+ Ph1

    t (9;22)

    Pathogenesis of Chronic Myelogenous Leukaemia

    bcr-abl 210 kDa

    tyrosine kinase activity

    constitutively active

    9q+ Ph1

  • The Gleevec Story:

    Bcr-Abl a Good Target

    A distinct chromosomal translocation the Philadelphia chromosome

    Bcr-Abl gene formed by juxtaposition of c-Abl oncogene on chromosome 9 with the Bcr oncogene on chromosome 22

    Resulting oncoprotein with elevated tyrosine kinase activity seen in 95% of patients with chronic myelogenous leukaemia (CML)

    A distinct drug target clearly differing in activity between normal and leukaemic cells

  • Gleevec: Chemical Optimisation

    Initial compound identified in high-throughput screen

    Promising lead-like properties high potential for chemical diversity

    Lead eventually to Gleevec (SAR studies aided by molecular modelling)

    Gleevec binds in ATP pocket, induces apoptosis in CML cells and causes dose-dependent inhibition of tumour growth in xenograft mouse models

    N

    N N

    H

    N

    Protein kinase Cinhibitor

    N

    N N

    H

    N

    Me

    N

    H

    O

    N

    N

    STI571 (Gleevec)

    (Glivec; Imatinib)

  • Gleevec: Mechanism of Action

    Initial compound identified in high-throughput screen

    Promising lead-like properties high potential for chemical diversity

    Lead eventually to Gleevec (SAR studies aided by molecular modelling)

    Gleevec binds in ATP pocket, induces apoptosis in CML cells and causes dose-dependent inhibition of tumour growth in xenograft mouse models

  • The Gleevec Story: Clinical Activity

    Outstanding clinical activity in CML patients, particularly in chronic phase of disease (95% response in Phase II human trials)

    Much lower response (29%) and high relapse rate in later stages of disease (blast crisis)

    Other tyrosine kinase receptor targets of Gleevec include c-kit in GastroIntestinal Stromal Tumours (GIST), and Platelet Derived

    Growth Factor (PDGF) receptor (variety of tumours)

    Drug resistance in late stage disease mutations in ATP binding pocket

    Novel agents - active in Gleevec-resistant disease:

    ( Nilotinib, Dasatinib. Weisberg et al, Nature Reviews cancer 7 p345 2007)

  • Point mutation: B-RAF

    - a single base is altered

    at position 1799 thymine is replaced by adenine

    - an altered codon

    - different amino acid

    Valine (V) is replaced by glutamate (E) at codon 600

    protein function

    Constitutive activation

    V600E mutation

    drives pathogenesis

    of > 60% Metastatic

    Melanoma

  • VEMURAFENIB

    V600E mutated BRAF inhibition

    August 2011: FDA approves Zelboraf (vemurafenib) and companion

    diagnostic test for BRAF mutation-positive metastatic

    melanoma, a deadly form of skin cancer.

  • VEMURAFENIB

    V600E mutated BRAF inhibition

    Mechanisms of resistance to vemurafenib

    Cancer cells may overexpress a cell surface protein PDGFRB creating an alternate survival pathway.

    NRAS mutation, reactivating the normal BRAF survival pathway

  • Vemurafenib vs Dacarbazine in metastatic melanoma patients

    Chapman et al NEJM 2011

  • 75% invasive breast cancers are classified ER+;

    65% are ER+ and PR+

    Treatment strategies for ER+ breast cancer:

    Inhibition of oestrogen synthesis

    Oestrogen receptor blockade

    Growth of ER+ breast tumours is stimulated by oestrogen.

    Breast Cancer UK statistics:

    In 2008 12,000 women and 70 men died from breast cancer

  • Targeting Oestrogen synthesis in ER+ Breast Cancer

    Aromatase inhibitors

    X X

  • Oestrogen receptor blockade

    Tamoxifen, Raloxifene

  • Anastrazole

    (Arimidex)

    Tamoxifen

    Treatment strategies for ER+ breast cancer:

    Inhibition of oestrogen synthesis Oestrogen receptor blockade

  • In 25-30% metastatic breast cancer patients the human

    epidermal growth factor receptor 2 - HER2 protein is

    overexpressed through amplification of the HER2 gene.

    HER2 signaling, particularly

    activation of downstream effector

    pathways including the

    Ras/Raf/MAPK and PI3K/Akt

    pathways, has been implicated in

    the pathogenesis of breast

    cancer.

    HER2+ overexpression is

    associated with a poorer

    prognosis than HER2- tumours.

    These cancers are less

    responsive to conventional

    chemotherapy and hormonal

    therapy.

  • Herceptin a humanised monoclonal antibody

    specifically blocks HER2 on the surface of breast

    cancer cells and turns off the permanently on signal for cell division.

    GFR

    Ras PI3K

    Raf AKT

    Mek mTOR

    Proliferation

    apoptosis resistance

    invasion, angiogenesis

    protein translation

    PTEN

  • Herceptin increases the survival

    time of women with advanced

    breast cancer.

  • Androgen-dependent prostate cancer:

    Inhibition of androgen synthesis:

    Critical role of CYP 17 in androgen synthesis

    Inhibition of CYP17 hydroxylase and lyase activities by Abiraterone

  • Treatment for Androgen-dependent prostate cancer:

    Abiraterone

    Inhibition of prostate cancer cell proliferation

  • Targeted therapy for the treatment of Non Small

    Cell Lung Cancer

    Background

    NSCLC most common form of lung cancer leading cause of cancer mortality Too advanced to be operable in ~50% patients First-line platinum agents give only modest increase in

    survival

    Limited efficacy and side effects of cytotoxic chemotherapy need for new therapies

    Aim

    To achieve cancer specific effects by targeting aberrant molecular pathways underlying tumour

    growth to improve efficacy and minimise side effects

  • Activation of EGFR is involved in NSCLC growth and progression:

    Inhibition of apoptosis Proliferation Angiogenesis Metastasis

    Lancet Oncology 2006; 7:499

  • Aberrant signalling important in development and

    progression of NSCLC

    EGFR expressed in up to 93% and overexpressed in ~45% of NSCLCs Level of EGFR expression correlates with poor prognosis and reduced

    survival

    good target

  • Lancet Oncology 2006; 7:499

    Targeting EGFR

    Monoclonal antibodies directed against extracellular ligand binding domain e.g. cetuximab (Erbitux) prevent ligand binding

    Small molecule selective inhibitors of intracellular TK domain e.g. gefitinib (Iressa; erlotinib (Tarceva) block autophosphorylation

    Downstream

    Signal

    Transduction

    Inhibited

  • EGFR Inhibitors: IressaTM and TarcevaTM

    Gefitinib Erlotinib

    N

    N

    N

    F

    Cl

    O

    MeO

    N

    OH

    ZD1839 (Iressa)

    N

    N

    N

    O

    O

    H

    OSI-774 (Tarceva)

    O

    O

    Synthetic small molecule receptor tyrosine kinase inhibitor

    Compete with ATP binding to intracellular TK domain of EGFR

    Inhibit receptor autophosphorylation

    Block downstream signal transduction

    Orally available

  • Tarceva: place in therapy

    Approved November 2004 for 2nd line tx of advanced NSCLC

    Antitumour activity and symptom relief (improved quality of life) in

    patients where standard chemotx

    has failed; mild adverse effects

    (skin rash, diarrhoea)

    Erlotinib Placebo

    End Points n = 488 n = 243 P value

    Progression-free survival (months)

    2.2 1.8 < 0.001

    Overall survival (months)

    6.7 4.7 < 0.001

    1-year survival (%) 31 22

  • NSCLC responders vs. non-responders to EGFRi

    Somatic mutations in the TK domain of EGFR correlates with subset of

    patients who are exquisitely sensitive:

    Women; never smokers; east Asian descent; adenocarcinoma;

    bronchoalveolar carcinoma.

    Tarceva

  • Efficacy of Gefitinib vs C/P in East Asian Patients

    with NSCLC

    Objective Response Rate (ORR) Gefitinib C/P P-value

    Intent-to-treat population

    (n = 609; 608) 43.0% 32.2%

  • Personalised Medicine Reduces Ineffective

    Treatment in Colon Cancer

  • KRAS mutation testing in metastatic colorectal cancer

    World J Gastroenterol. 2012 October 7; 18(37): 51715180.

    Published online 2012 October 7. doi: 10.3748/wjg.v18.i37.5171

    PMCID: PMC3468848

    kras Testing

    Do Not Treat

    Treat with Erbitux

    Treat with Erbitux

    Treatment

    Success

  • In situ carcinoma: undetectable microscopic mass of tumour cells

    Tumour cells secrete molecules

    (e.g. vascular endothelial growth

    factor, VEGF)

    Signalling activates genes/ protein

    synthesis in normal host tissue

    encouraging growth of new blood

    vessels to the tumour

    Judah Folkman 1933-2008 Founder anti-angiogenic therapy

    One of the Hallmarks of Cancer: sustained Angiogenesis

    promotes: Growth of primary tumour Metastasis

  • Hypoxia, transforming (oncogenic,

    tumour suppressor) mutation:

    HER2,ras, P53, PTEN, PI3K/AKT, NO, bFGF, Il-8, EGF, IGF-1, PDGF, COX-2, H2O2

    VEGF

    synthesis/release

    P

    P

    P

    P

    Binding of VEGF to

    VEGFR

    VEGFR activation

    Survival, proliferation,

    migration

    ANGIOGENESIS

    Role of VEGF in Angiogenesis / Tumourigenesis

    Potent mitogen for vascular

    endothelial cells

    Mediates the secretion and activation of enzymes which

    degrade the extracellular matrix

    e.g. matrix metalloproteinases,

    collagenase

    Survival factor for endothelial cells through inhibition of

    apoptosis

    Modulates endothelial cell migration

    to sites of angiogenesis

    Vascular maintenance

    Lymphangiogenesis

    VEGF

  • VEGF a good target ?

    Key promoter of metastasis and central role in tumour growth

    VEGF expression is increased in the majority of cancers examined to date

    Overexpression correlates with risk of recurrence and poor prognosis

    Drugs targeting VEGF activity at endothelial cells do not need to penetrate tumours

    Drug resistance less likely than with traditional chemotherapeutics: greater genetic stability of endothelial cell compared to cancer cell

  • Targeting VEGF(R)

    VEGF/VEGFR blockade by

    monoclonal antibodies

    Inhibition of receptor

    signalling by small molecule

    tyrosine kinase inhibitors

    VEGF

    Kinase

    inhibitors Anti-VEGFR-1/2

    VEGF receptor

    Aptamer

    Anti-VEGF

    SiRNA

    decoy receptor

    Ribozyme targeting

    VEGFR mRNA

  • How to target VEGF

    BEVACIZUMAB Avastin;

    Humanised neutralising IgG

    monoclonal antibody directed

    against VEGF

    Binds all VEGF isoforms with

    high affinity and blocks their

    binding to the receptors.

    VEGF

    Kinase

    inhibitors Anti-VEGFR-1/2

    VEGF receptor

    Aptamer

    Anti-VEGF

    SiRNA

    decoy receptor

    Ribozyme targeting

    VEGFR mRNA

  • Place in therapy of Bevacizumab (Avastin)

    Metastatic colorectal cancer (mCRC)

    The addition of bevacizumab to 5-FU and irinotecan regime

    increased survival time by ~ 5

    months (30%) (20.3 months

    versus 15.6 months; p < 0.001)

    Approved (2004) in combination with first-line 5-FU-based

    chemotherapy regimens

    Bevacizumab plus chemotherapy improved survival in metastatic non-small cell

    lung cancer in a randomized clinical trial*

  • Atkins et al. Nature Reviews Drug Discovery 5, 279280 (April 2006) | doi:10.1038/nrd2012

    Small molecule inhibitor of VEGFR: Sunitinib

  • Results of Phase III trial of Sunitinib in glivec refractory GIST patients.

    (G Demetri 2006).

    Time to tumour progression

  • Sunitinib (Sutent) small molecule inhibitor of

    VEGFR

    Orally available small-molecule inhibitor of VEGFR 1, 2 and 3, PDGFR , , c-kit and flt-3 rich and diverse pharmacology

    Phase II trials completed in cytokine-refractory metastatic renal cell cancer

    Unusually high rate of objective tumour responses detected - 40%;

    stable disease - 27%

    Acquired resistance: rapid enhanced migratory and invasive capacity

  • Targeted therapies for solid tumours?

    Gleevec success in CML where a single genetic alteration underpins disease pathogenesis (bcr-abl)

    Common solid tumours do not have one defined genetic alteration driving the neoplastic process

    Clinically heterogeneous disease

    Acquired resistance: Target mutation (bcr-abl),

    Elevated target expression (BRAF V600E)

    Activation / upregulation of alternative signalling pathways (EGFR MET),

    Acquired oncogenic mutations in complementary signal networks (EGFR Ras)

    Enhanced cancer cell migration / invasion capacity

  • Mutation/loss of

    FAP gene

    APC

    DNA

    methylation

    KRas

    mutation

    Loss of

    P53

    Acquisition of

    multiple genetic

    mutations

    Solid Tumourigenesis

  • Targeting Solid Tumours

    Combination of

    molecularly-targeted agents with cytotoxic /

    radiotherapy

    Erbitux (cetuximab)

    targets EGFR with

    Irinotecan advanced colorectal cancer

    Herceptin (Trastuzumab) targets HER2 protein with

    paclitaxel HER2+ metastatic breast cancer

    Avastin targets VEGF with paclitaxel or

    carboplatin in NSCLC

    Dual / multiple TK inhibitors

    Zactima Sunitinib Sorafenib

    Lapatinib: EGFR / HER2 GSK

    small molecule Herceptin refractory breast, lung cancer

    Capecitabine 5FU prodrug

  • The Phase III registration trial: Combination

    with chemotherapy

    Use of chemotherapy plus a monoclonal antibody against HER2 for

    metastatic breast cancer that overexpresses HER2

    Few studies of metastatic breast cancer demonstrated a

    survival advantage of this magnitude.

    Slamon DJ et al. N Engl J Med 2001; 344:783-792.

    Prior anthracyclines

    Herceptin + paclitaxel (n=92)

    Paclitaxel (n=96)

  • Significant improvement in progression-free

    survival (primary endpoint)

  • Summaryso far

    The Molecular Mechanisms underpinning tumourigenesis are

    Numerous,

    Complex,

    Not fully understood

    Target aberrant proteins

    Small molecules

    Antibodies

    Resistance

    Combine cytotoxic and molecular targeted agents

    to defeat cancer

  • Combination Therapy: Lung Cancer

  • Combination Therapy: Lung Cancer

    Small cell lung cancer - chemotherapy (responds well; also radiotherapy)

    Non small cell lung cancer (NSCLC)

    - surgery, radiotherapy,

    Biological therapy: EGFR inhibitors

    Chemotherapy:

    Cisplatin / carboplatin -

    Etoposide -

    Doxorubicin -

    Topotecan -

    Vincristine -

    Gemcitabine -

  • Combination Therapy: Lung Cancer

    Small cell lung cancer - chemotherapy (responds well; also radiotherapy)

    Non small cell lung cancer (NSCLC)

    - surgery, radiotherapy,

    Biological therapy: EGFR inhibitors - Tarceva, Iressa

    Chemotherapy:

    Cisplatin / carboplatin - cross links DNA

    Etoposide - Topo II inhibitor

    Doxorubicin - DNA intercalator (heart complications)

    Topotecan - Topo I inhibitor (camptothecin derivative)

    Vincristine - antimicrotubule agent

    Gemcitabine - antipyrimidine

  • Combination Therapy:

    Colorectal (Bowel) Cancer

  • Combination Therapy:

    Colorectal (Bowel) Cancer

    Surgery (8/10)

    Radiotherapy

    Chemotherapy

    5-fluorouracil (5-FU) -

    Oxaliplatin -

    Irinotecan -

    Biological therapy

    Erbitux; Iressa -

  • Combination Therapy:

    Colorectal (Bowel) Cancer

    Surgery (8/10)

    Radiotherapy

    Chemotherapy

    5-fluorouracil (5-FU) - antipyrimidine

    Oxaliplatin - platinum DNA crosslinker

    Irinotecan - topoisomerase 1 inhibitor

    Biological therapy

    Erbitux, Iressa - EGFR inhibitors