• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/740

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

740 Cards in this Set

  • Front
  • Back
What does it mean when a tumor is well-differentiated?
It resembles the surrounding tissue and has a good prognosis
What transition will cancer cells undergo during metastasis?
Epithelial --> Mesenchymal
What happens to cell arrangement in cancer progression?
Atypical hyperplasia: fairly normal arrangement; CIS: random alignment of cells; Invasive: loss of cell-cell contacts – these changes occur due to disruption of actin filaments
What can necrosis in a tumor be associated with?
A malignant tumor that has outgrown its blood supply
What is the earliest stage that can be detected by mammography?
Carcinoma in situ
What is the progression from a normal breast to metastatic breast cancer?
Normal breast --> atypical hyperplasia --> carcinoma in situ (stages 1-4/comedo) --> invasive breast cancer --> metastatic breast cancer
What is the most common serum tumor marker used?
PSA for prostate cancer
What are the most common cancers in the US?
Lung > breast/prostate > colon/rectum
What differentiates between carcinoma, sarcoma, and teratoma?
Carcinoma: derived from epithelial cells; Sarcoma: derived from mesenchymal cells; Teratoma: derived from germ cells
What type of cancer is more common in WV when compared to other states?
Cervical
What are common metastatic sites?
Melanoma --> Lungs; Prostate --> Bone; Colon --> Liver/Pancreas
What is the most commonly mutated protein in cancers and what is this protein involved with?
p53 – gatekeeper protein that provides a checkpoint in the cell cycle; high levels indicate dysfunction
What is the first Cyclin to appear in G1 and what chain of events follows its production?
Cyclin D1 --> it binds to cdk4 forming a complex that phosphorylates Retinoblastoma susceptibility protein (RB) --> RB phosphorylation causes its release from the transcription factor EF2 --> this activates EF2, allowing it to transcribe genes required for initiation of progression into S-phase
What is the Abl gene and how does it cause cancer?
It codes for a non-receptor tyrosine kinase which shuttles between the nucleus where it regulates gene transcription and the cytoplasm where it regulates the actin cytoskeleton and cell movement; Abl on chromosome 9 fuses with BCR on chromosome 22 to form the Philadelphia chromosome – Imatinib (Gleevac) diminishes Abl activity
How does Ras function in tumor cells?
A mutation causes Ras to be unable to self-hydrolyze its GTP to GDP, in effect keeping it in its activated state continuously; in this state it activates gene transcription through the MAP kinase pathway
How does overexpression of growth factors often function in tumor cells?
In conjunction with overexpression of growth factor receptors, allowing for an autocrine loop
What defines growth factor receptors ascribable as oncogenes and what are some examples?
Typically, transmembrane tyrosine kinase receptors that dimerize and trans-phosphorylate/activate upon stimulation by a ligand; Examples: EGFR, HER2/neu (Erb-2), FGFR, HGFT (Met)
What defines oncogenes and what are the classic examples?
An oncogene refers to a normal cellular gene (proto-oncogene) that when activated or overexpressed produces a dominant phenotype in spite of the presence of the normal allele;
Examples: growth factor receptors, growth factors, Ras, Abl, Src, Myc
How do cancer cells increase their Cyclin levels?
By amplification of their Cyclin genes
Are most tumors that present clinically homogenous or heterogenous and what does this mean?
Heterogenous: the progeny of the initial cancer cell can acquire different genetic damage over time
What essential physiological changes are required for neoplasia?
Tumor cells lack a requirement for growth signals; They are refractory to growth-inhibition signals (e.g. TGF-β); They are resistant to apoptosis (via inactivation of p53); They have defective DNA repair; They can replicate indefinitely; They initiated and sustain their own angiogenesis (secrete VEGF); Most tumors have the ability to invade and metastasize
What compounds regulate the cell cycle and by what mechanism?
G1: Cyclin D/cdk4; S: Cyclin E/cdk2; G2: Cyclin A/cdk2; M: Cyclin B/cdk1;
CDKs are constitutively present in the cell, but inactive --> Specific cyclins that activate the CDKs are produced for each phase of the cell cycle by complexing with them --> These complexes allow progression of the cell cycle
How does RB cause cancer?
By failing to suppress gene transcription; only happens in individuals who are homozygous for the mutated allele (Knudson’s two-hit hypothesis: need two mutations to cause RB); in people who are heterozygous for the RB mutation a “loss of heterozygosity” (LOH) will lead to RB
What are BRCA1/2 involved in?
DNA repair mechanisms (fixes double-stranded breaks) – BRCA1/2 are involved in breast cancer
How can changes in the BCL-2 pathway lead to cancer?
BCL-2 is an anti-apoptotic protein that protects mitochondria from the influence of death pathways; In B-cell follicular lymphomas BCL-2 is translocated from chromosome 18 to 14 (same place where Myc translocation takes place), leading to uncontrolled transcription of BCL-2 thus preventing lymphoid cells from dying
In what genetic disease are APC mutations found?
Familial Adenomatous Polyposis (FAP) --> leads to benign and malignant colon polyps
What is the mechanism by which absence of APC leads to cancer?
APC functions by destroying excess β-catenin; β-catenin is normally bound to E-cadherin, but translocates to the nucleus upon WNT stimulation of frizzled --> in the nucleus β-catenin activates the transcription factor TCF which in turn increases Myc and Cyclin D1 levels;
In what syndrome does inheritance of a single mutated p53 gene cause a 25x greater risk for developing cancer in variety of locations by age 50?
Li-Fraumeni syndrome
What is the most important gene involved in cancer and how does it function?
p53: normally prevents the propagation of cells with genetic damage; it does this by first halting the cell cycle (p53 is activated by ATM kinase after DNA damage has occurred) via p21 which inhibits CDKs --> this allows DNA repair enzymes (such as GADD45 induced by p53) to fix the damage --> if the damage is fixed p53 is degraded, if not, p53 induces apoptosis via the Bax gene
What is Src and how does it cause cancer?
It is a non-receptor tyrosine kinase that regulates cell cycle progression and the actin cytoskeleton; it also predisposes cells to undergo the epithelial-mesenchymal transition (EMT) seen in metastasis by altering the cytoskeleton and producing metalloproteinases for ECM degradation; Treated with Dasatinib and AZD0530 (clinical trials)
What is the function of Myc and how does it cause cancer?
It is a nuclear transcription factor involved in the cell cycle (Cyclin D2/histone deacetylation), cell movement, and cell adhesion; in tumor cells Myc is often upregulated – This is the cause Burkitt’s lymphoma (B-cell tumor) as a result of a chromosome 8 to 14 translocation (similar to BCR/Abl)
What are some common tumor suppression genes?
RB, p53, APC/β-catenin
What is meant by the multi-hit hypothesis for carcinogenesis?
Multiple genetic events are required to form a full blown tumor: mutations in DNA-repair enzymes (caretaker pathway) --> genomic instability --> mutations in tumor suppressor genes (gatekeeper pathway) --> activating mutations in oncogenes --> tumor cell
What happens to tumor cells that lose expression of MHC-I?
They become more susceptible to killing by NK cells (although NK cells are often not potent enough to cause complete destruction of the tumor – needs to be followed by a T cell response)
What can tumor cells be transfected with in order to enhance tumor immunogenicity?
B7 or GM-CSF
How are monoclonal antibodies used in tumor cell destruction?
By themselves, in binding to the tumor cells and activating NK cells; In a conjugated form with a toxin, causing ingestion of the toxin into the tumor cell; In a conjugated form with a radionuclide, causing death of the tumor cell and its neighbors by radiation
How do heat-shock proteins enhance the immune response to tumors?
They bind tumor antigens and carry them to dendritic cells where they can be degraded and presented on both MHC-I and MHC-II for T cell activation
How can epithelial tumor cells evade tumor-infiltrating lymphocytes?
By cleaving off their own surface receptor, MIC --> soluble MIC can then bind to NKG2D on tumor-infiltrating lymphocytes, causing the internalization of these NKG2D receptors --> this reduces the levels of NKG2D on the tumor-infiltrating lymphocytes, making them less effective
How does the melanoma peptide vaccine (MAGE-1/-3) cause tumor destruction?
It induces anti-melanoma CD8+ CTLs
By what mechanisms can tumor cells be recognized by T cells?
Mutated peptide is presented by MHC-I; Reactivated embryonic proteins are presented by MHC-I; Overexpression of normal cell proteins causes changes in density of self-peptide presentation
Which immune cells are the most effective in controlling tumor cells?
CD8+ CTLs
By what mechanisms can tumors escape immune recognition?
Tumor cells have low immunogenicity: lack of MHC, adhesion and co-stimulatory molecules; Antigenic modulation: endocytosis of antibodies against the tumor cells; Tumor-induced immune suppression: factors secreted by the tumor inhibit T cells
What are two examples of tumors that are not neoplastic and what characterizes each?
Choristoma: ectopic rest of normal tissue; Hamartoma: a mass of disorganized but mature specialized cells indigenous to a particular site – i.e. a choristoma is made up of elements not normally found at that site, whereas a hamartoma is made up of tissue that is normally found at that site
What characterizes benign neoplastic lesions?
Cohesive, expansile mass; remain localized to their site of origin; well-differentiated; slow growth
What characterizes malignant neoplastic lesions?
Invade adjacent structures/spread to distant sites; may be poorly differentiated or undifferentiated; may grow rapidly
What is meant by the term direct seeding?
Penetration of a tumor into a natural open space (e.g. peritoneal, pleural, pericardial cavities)
What is a cancer called that has not invaded surrounding tissues yet?
Dysplasia (carcinoma-in-situ)
What is it called when one adult cell type is replaced by another due to environmental stress?
Metaplasia (makes tissue much more susceptible to malignant transformation)
What are some key morphologic features of malignant neoplasms?
Pleomorphism (i.e. variation in size/shape); Hyperchromasia, increased nuclear/cytoplasm ratio, large nucleoli; Mitotic figures; Loss of cell polarity; Tumor giant cells; Necrosis
What defines the term anaplasia?
Lack of differentiation (i.e. it does not resemble comparable normal cells)
What defines the term desmoplasia?
The formation of an abundant collagenous stroma in response to tumor growth and invasion
What is meant by the term clonality?
The entire population of cells within a neoplasm arises from a single cell
How does the spread of epithelial vs. mesenchymal malignancies differ, in general?
Epithelial malignancies spread to lymphatics; Mesenchymal malignancies to the blood supply
What suffix do embryonal tumors have associated with them?
-blastoma (small, round, blue-cell tumor of children)
What is a tumor called that originates from pluripotential stem cells and can differentiate into any type of tissue?
Teratoma
What are the terms used for benign and malignant mesenchymal neoplasms?
Benign: -oma; Malignant: sarcoma
What are the terms used for benign and malignant epithelial neoplasms?
Benign: -oma; Malignant: carcinoma (e.g. adenoma vs. adenocarcinoma)
What differentiates between T2 and T3, as well as N2 and N3?
T2: larger, more invasive within the primary organ site; T3: larger/invasive beyond the margins of the primary organ site; N2: extensive regional lymph node involvement; N3: more distant lymph node involvement
What are examples of serum tumor markers used in molecular diagnosis?
Carcinoembryonic antigen (CEA): carcinomas and benign disorders; Alpha-fetoprotein: hepatocellular carcinoma and germ cell tumors; Human chorionic gonadotropin: trophoblastic and germ cell tumors; CA125: ovarian tumors; CA19-9: pancreatic and colonic cancers
What are examples of prognostic/therapeutic molecules that affect breast cancer?
Estrogen/progesterone receptors (receptor positive tumors have a better prognosis due to susceptibility to antiestrogen therapy); Overexpression of c-erb-B2 or HER2/neu protein (poor diagnosis)
What are PSA and TTF1 used for?
Determining site of origin for metastatic tumors; PSA: prostate; TTF1: lung, thyroid
What antibodies are used to identify carcinomas vs. sarcomas?
Carcinomas: Cytokeratin; Sarcoma: Vimentin; other antibodies used: lymphoma/CD45, glioma/GFAP
What is immunohistochemistry used for in cancer?
Categorization of undifferentiated malignant neoplasms, as well as leukemias/lymphomas; Determining the site of origin of metastatic tumors; Detection of prognostic/therapeutic molecules
What are two techniques used in cytologic diagnosis of cancer?
Fine-needle aspiration: used for readily palpable lesions (thyroid, lymph node); Cytologic smears (Pap smear, urine)
What differentiates between grading and staging?
Grade: level of differentiation; Stage: extent of spread
What is measured in the TNM system?
T: primary tumor size/extent of invasion; N: regional lymph node involvement; M: Metastases
What is a retinoblastoma?
Small, round, blue-cell tumor that occurs in the retina, usually in children, due to a defect in RB
What is one of the most common syndromes that involve DNA repair mechanisms associated with mismatch repair and how can this be tested for?
Hereditary nonpolyposis colorectal cancer (HPNCC) – tested for by looking at microsatellite instability (changes of simple sequence repeats in DNA)
What does the loss of the VHL gene on chromosome 3 lead to?
von Hippel Lindau Syndrome: hemangioblastomas of the brain/retina and renal cell carcinomas;
also pheochromocytomas, microcystic adenomas of the pancreas, cystic lesions of the epididymis, and endolymphatic sac tumors
On what chromosomes are NF-1 and NF-2 found, respectively?
NF-1: 17 (as in the # of letters in “von Recklinghausen”); NF-2: 22 (with bilateral eight nerve schwannomas)
A mutation in what gene causes Familial Adenomatous Polyposis?
APC gene on chromosome 5 – APC down-regulates β-catenin (involved in regulation of transcription factors and cell cycle genes); leads to innumerable polypoid adenomas of the colon
What syndrome is associated with a germ-line mutation of p53?
Li-Fraumeni syndrome (leads to multiple carcinomas and sarcomas)
What is the most common target for genetic alteration in human tumors and what chromosome is it found on?
p53 on chromosome 17
How are familial cancers most often inherited and what are they mostly associated with?
Autosomal dominant – defects in tumor suppressor genes
On what chromosome is RB located and what transcription factor does it regulate?
Chromosome 13; regulates E2F transcription factors
Besides the increased risk for retinoblastomas, what other cancer is associated with RB?
Osteosarcomas
What symptoms are associated with cancer cachexia (wasting syndrome) and what is thought to cause it?
Loss of body mass, weakness, anorexia, anemia – thought to be caused by production of TNF and other cytokines)
What are some examples of neuromyopathic paraneoplastic syndromes?
Peripheral neuropathies; Cerebellar cortical degeneration (Purkinje cell antibodies = ataxia); Limbic encephalitis; Lambert-Eaton myasthenic syndrome (antibodies to presynaptic terminals at neuromuscular junctions)
What paraneoplastic syndrome is caused by 5-HIAA?
Carcinoid Syndrome (seen with carcinoid tumors): flushing, diarrhea, heart disease
What is Trousseau Syndrome?
A hypercoagulable state associated with various carcinomas
What paraneoplastic syndrome is seen in renal cell carcinomas?
Polycythemia (~Erythropoietin)
What are examples of paraneoplastic syndromes that are seen in lung cancer?
Oat cell (small cell): Cushing’s syndrome (~ACTH), Hyponatremia (~SIADH); Squamous cell: Hypercalcemia (~Parathormone)
What is meant by paraneoplastic syndromes?
Symptom complexes in cancer-bearing patients that cannot be explained by the local or distant spread of the tumor nor by the elaboration of hormones indigenous to the tumor tissue
What is the defect in Xeroderma Pigmentosum?
A defect in nucleotide excision repair – leads to squamous carcinoma of the skin, especially due to UVB rays (since they cause the formation of pyrimidine dimers)
What DNA repair defect causes increased susceptibility to DNA damage by ionizing radiation, oxygen free radicals, and DNA cross-linking agents?
Defects in DNA repair by homologous recombination – e.g. Ataxia-telangiectasia, Bloom syndrome, Fanconi anemia
What nonhereditary predisposing conditions are associated with cancer?
Inflammatory diseases: ulcerative colitis, Crohn’s disease, Helicobacter pylori gastritis, viral hepatitis, chronic pancreatitis; Hyperplasia/Dysplasia/Metaplasia; Hepatic cirrhosis; Pernicious anemia; Solar keratosis; Leukoplakia (squamous hyperplasia of oral mucosa)
What is hyperpigmentation of the skin called that is classified as a paraneoplastic syndrome?
Acanthosis nigricans
What paraneoplastic syndrome is associated with lung cancer (bronchogenic carcinoma)?
Hypertrophic osteoarthropathy
What are the most common types of cancer in males and females, respectively?
Males: prostate>lung>colon/rectal; Females: breast>lung>colon/rectal
What does Bavacizumab affect?
Angiogenesis
What cancer inducers are affected by Imatinib?
C-kit, BCR-Abl, Platelet Derived Growth Factor (PDGF)
Which drugs function at the level of the HER receptor (EGFR)?
Cetuximab, Trastuzamab, Lapatinib, Erlotinib
What does HER2 overexpression lead to?
HER2 dimerizes with HER1,3,4 and provides signaling, leading to increased cell proliferation, increased cell migration, and resistance to apoptosis – overexpression of HER 2 will increase all these aspects and decrease patient survival rates
What are the screening recommendations for breast cancer, colon cancer, cervical cancer, and prostate cancer?
Breast: age 40; Colon: age 50; Cervical: 3 years after vaginal intercourse but no later than age 21; Prostate: age 50 (or 45 for high risk individuals – i.e. family history or African American)
What is the major target for anti-cancer drugs?
DNA: precursors, direct attack, replication enzymes
How is extravasation caused in regard to anti-cancer drugs?
When the anti-cancer drug leaks out or settles at the IV injection site, it can act as a vesicant and cause severe skin irritation and destruction
What can be a complication of mucositis/stomatitis associated with anti-cancer drugs?
Due to the myelosuppression associated with anti-cancer drugs, patients can develop fungal infections in their upper GI tract, thus worsening their mucositis/stomatitis
What is done about the nausea and vomiting associated with anti-cancer drugs?
It is treated with medication before the actual onset of symptoms since it is harder to beat once the patient is nauseous/vomiting
What is the lowest value of blood counts after chemotherapy called?
Nadir
What common toxicities are associated with anti-cancer drugs?
Tumor lysis syndrome: tumor cell contents are released causing hyperkalemia/ hyperphophatemia/increased uric acid (more common in leukemias/lymphomas); Myelosuppression; Nausea/Vomiting; Mucositis; Alopecia; Extravasation; Infertility; Secondary malignancies;
What are the major problems with many anti-cancer drugs?
The anti-cancer drugs not only affect cancer cells, but also normal cells that are rapidly dividing
What is meant by Gompertzian growth of cancers?
The initial growth phase is linear on log scale (i.e. high growth fraction/short doubling times); this is followed by a plateau growth phase (i.e. lower growth fraction/longer doubling times) – plateau phase possibly due to vascular needs that can’t be met
For what cancer size is immunotherapy beneficent?
Small tumor size (i.e. <105 cells)
What are the primary treatment options for cancers?
Surgery, radiation, chemotherapy, biotherapy
How can anti-cancer drugs cause secondary malignancies?
Besides attacking the cancer, they also act as mutagens, thus being able to cause secondary malignancies
What is an important mechanism by which antibiotics work?
They intercalate DNA, thus disrupting its normal functioning
What are the pharmacokinetics associated with CTX?
It is a pro-drug, activated by CYP450 to a phosphoramide mustard; CTX is detoxified by aldehyde dehydrogenase (ALDH) to a carboxy compound – besides being activated by CYP450, it is also dechloroethylated into dechloroethyl (neurotoxic) and chloroacetaldehyde (nephrotoxic)
What is the mechanism of action of Cyclophosphamide (CTX) and what are its indications?
Bifunctional alkylation of DNA, producing cross-links; Used as an anti-cancer drug and as an immunosuppressant
What are the alkylating agents used in chemotherapy?
Mechlorethamine, Cyclophosphamide, Ifosfamide, Melphalan, Chlorambucil, Busulfan, Carmustine, Lomustine, Streptozocin, Thiotepa, Dacarbazine, Procarbazine (non-classical)
What differentiates adjuvant therapy from neo-adjuvant therapy?
Adjuvant therapy: giving chemotherapy after surgery in order to prevent relapse;
Neo-adjuvant therapy: giving chemotherapy prior to surgery in order to reduce tumor burden
What factors are cycle and dose rate determinants?
Doubling time of the tumor; Time to recover from toxicity; Dosage regimen (low/high, intermittent/continuous); Subjective manipulation; Target (e.g. continuous for inhibiting angiogenesis and intermittent for drugs with cytotoxic effects)
What anti-cancer drugs affect the winding/unwinding of DNA?
Topoisomerase inhibitors
What are the three classification mechanisms used for anti-cancer drugs?
Chemical (not very useful); Cell cycle dependency: cycle independent (nonspecific), cycle specific (proliferation dependent), phase specific (only affects cells in a specific cell cycle phase); Mechanistic classifications (most useful clinically)
By what mechanism do alkylating agents work?
They covalently bind to the bases on DNA, causing mispairing/mutations/cross-linking/breaks
How do the antimetabolite anti-cancer drugs function?
They are structural analogs of endogenous precursor molecules for DNA/RNA biosynthesis; they interact with enzymes involved in nucleic acid biosynthesis, thus inhibiting predominantly DNA biosynthesis
By what important mechanism do cancer cells become resistant to anti-cancer drugs?
Alterations in drug transport (changes involving the p-glycoprotein transporter – MDR)
What is the mechanism of action of Carboplatin and Cisplatin and what are their respective pharmacokinetics?
They bind covalently to DNA; Carboplatin PK: 70% urine excretion; Cisplatin PK: inactivated by avid protein binding
What is the mechanism of action of MTX?
Inhibition of dihydrofolate reductase --> partially depletes reduced (activated) folates necessary for DNA synthesis; Metabolites inhibit purine (via AICAR) and thymidylate (via dTMP) biosynthesis
What important toxicities are associated with Ara-C and what is its main indication?
Myelosuppression; Mucositis; CNS and chest pain (mainly in elderly) – Ara-C is used mainly in leukemias
What is the mechanism of action of Cytarabine (Ara-C) and what are its pharmacokinetics?
MOA: inhibits DNA polymerase, incorporated into DNA, terminates DNA chain elongation;
PK: inactivated by cytidine deaminase; very short half-life (~10 min.)
What antimetabolites are used in chemotherapy?
Cytarabine (Ara-C), Fludarabine, Gemcitabine, Pentostatin, Cladribine, 6-MP, MTX, Pemetrexed, 5-FU, Capecitabine,
What is special about the dosing calculation for Carboplatin?
It is based on creatinine clearance
What differentiates the toxicities associated with Carboplatin and Cisplatin?
Carboplatin: myelosuppression, nausea/vomiting (n/v); Cisplatin: nephrotoxicity (incl. reduced GFR), ototoxicity, n/v – Cisplatin nephrotoxic/ototoxic effects are possibly irreversible
What are the DNA binding agents used in chemotherapy?
Carboplatin, Cisplatin, Oxaliplatin
What important toxicities are associated with CTX?
Myelosuppression; Immunosuppression; SIADH; Bladder toxicity (must keep pt hydrated)
What toxicities are associated with MTX?
Myelosuppression; Mucositis; Renal obstruction (prophylactic treatment with urinary alkalinization and hydration) – also, accumulation of MTX in third-space fluids (i.e. ascites or pleural effusions) prolongs its half-life
What compound is given after high-dose MTX in order to “rescue” cells from toxicity?
Reduced folate
What are risk factors that predispose to B-cell lymphomas?
Primary immunodeficiency syndrome; Autoimmune disorders; Acquired immunodeficiency disorders; Viral associations; Bacteria immune response associations
What are the two different subtypes of CLL/SLL in regard to the stage at which the disease takes place?
Naïve: takes place at the naïve B cell stage, lack somatic mutations, worse survival (~3 years);
Post-germinal center: takes place at the post-germinal center B cell stage, associated with memory B cells, possess somatic mutations, better survival (~7 years)
What is the immunophenotype associated with CLL/SLL?
Positive for CD5, CD19, CD20, CD22, CD23, occasional CD38 (worse prognosis);
Negative for CD10, Cyclin-D1
What is the difference between Chronic Lymphocytic Leukemia (CLL) and Small Lymphocytic Lymphoma (SLL)?
The majority of patients have CLL: a marrow disease in which blood with small round lymphocytes and smudge/basket cells are found; The minority has SLL: a spleen disease (white pulp) in which lymph node diffuse effacement with proliferation centers and paraimmunoblasts are seen – includes extranodal sites: skin, breast, ocular
What characterizes the patient with CLL/SLL?
Usually asymptomatic although fatigue, autoimmune hemolytic anemia, and infections are possible; also lymphadenopathy and hepatosplenomegaly; >50 years of age; 2:1 male:female
What are the three categories of outcomes regarding B-cell lymphomas?
Indolent (incurable, median survival >5 years): CLL/SLL, Follicular
Slowly aggressive (incurable, median survival = 3 years): Mantle cell
Aggressive (potentially curable): Diffuse large B-cell, Burkitt, Lymphoblastic
What viral and bacterial infections are associated with B-cell lymphomas?
EBV: Burkitt lymphoma, post-transplant lymphoproliferative disorders
Human herpes 8 – Kaposi sarcoma herpes virus: primary effusion lymphoma
Hepatitis C: Lymphoplasmacytic lymphoma with type II cryoglobulinemia
Helicobacter pylori: Extranodal marginal zone lymphoma
What important CD markers are found in each of the three layers of the germinal centers?
Pale center: CD10+; Mantle zone: CD5+; Marginal zone: CD10-/CD5-
In hematopathology, what are the most common reasons for polyclonality and monoclonality, respectively?
Polyclonality: reactive, infectious causes; Monoclonality: malignant causes (although it can be autoimmune) – Autonomy refers to growth/proliferation outside of normal regulatory pathways
What chromosome translocations have been identified, in regard to B-cell lymphomas, which place oncogenes under Ig heavy chain gene promoter control on chromosome 14?
Mantle cell lymphoma, t(11;14): overexpression of Cyclin-D1
Follicular lymphoma, t(14;18): overexpression of BCL-2
Burkitt lymphoma, t(8;14): overexpression of c-MYC
What immunophenotype is associated with MCL?
Positive for CD5, Cyclin-D1 (sets it apart from CLL/SLL!), CD19, CD20, CD22;
Negative for CD10, BCL-6, CD23
What do 25-35% of Fl cases transform into?
Diffuse Large Cell Lymphoma
What is the immunophenotype of FL?
Positive for BCL-6, CD10, CD19, CD20, CD22, and BCL-2: due to t(14;18)!
Negative for CD5 and Cyclin-D1
How is FL graded?
By counting centrocytes (lymphocyte with cleaved nucleus) and centroblasts (lymphocyte with large non-cleaved nucleus; counts are done per 40x high power field (HPF) –
Grade I: 0-5 centroblasts/HPF; Grade II: 6-15/HPF; Grade III: >15/HPF
What are the classifications associated with follicle patterns in FL?
Follicular (closest to normal lymph node morphology); Follicular and Diffuse; Minimally Follicular
What are the differences between adult and pediatric Follicular Lymphoma (FL)?
Adult: female>male, asymptomatic with large nodes; Pediatric: rare, male>>female, head and neck involvement, high-grade lymphoma, better prognosis
What genetics are associated with MCL?
The defect is almost always at the naïve B-cell stage; t(11;14) present: links IgH to Cyclin-D1; BCL-2 and MYC rearrangements are absent
What is the prognosis for CLL/SLL?
It is indolent and noncurable – Transformation can lead to Diffuse Large Cell Lymphoma (3%, known as Richter Syndrome); transformation can also lead to Paraimmunoblastic type and Hodgkin disease
What sites in the body are involved in Mantle Cell Lymphoma (MCL)?
Blood: small irregular lymphocytes; Bone marrow; Lymph nodes: nodular/diffuse patterns, proliferation centers absent (as opposed to CLL/SLL!); Spleen disease (white pulp); Extranodal disease
What two specific extranodal diseases are seen with MCL and why?
Lymphomatous Polyposis (GI tract); Waldeyer’s ring involvement (throat) – affected because they express the α4β7 homing receptor
As a lymphoma becomes more aggressive, what happens to the sites of involvement?
They become more extensive (i.e. they are more likely to affect other organs)
What immunophenotype is associated with Burkitt’s lymphoma?
Positive for CD10, BCL-6, CD19, CD20, CD22; Negative for CD5, BCL-1, CD23, CD34, TdT
What morphological finds are associated with Burkitt’s lymphoma?
Monotonous diffuse infiltrates of medium sized blasts with sharp lipid vacuoles; High mitotic and apoptotic counts; Starry-sky appearance
What extranodal sites are commonly involved in Burkitt’s lymphoma?
Endemic: jaw, facial bones; Sporadic: abdominal mass, breasts (in pre-adolescent girl); Immunodeficiency: lymph nodes and marrow – growth of these sites is extremely rapid
What are the three clinical variants of Burkitt’s lymphoma?
Sporadic: world-wide, children and young adults; Endemic: equatorial Africa, 4-7 years of age, virtually all EBV positive; Immunodeficiency associated: HIV/transplantation – EBV is a risk factor for Burkitt’s lymphoma
What are the two major classifications of molecular profiling for Diffuse Large B-Cell Lymphoma and what is their prognostic significance?
Germinal center type: better prognosis; Activated blood B-cell type: worse prognosis
What immunophenotype is associated with Diffuse Large B-Cell Lymphoma?
Positive for CD19, CD20, CD22, BCL-6; Negative for CD10, CD5, BCL-1
What is the difference between primary and secondary Diffuse Large B-Cell Lymphoma?
Primary: de novo, can happen in immunocompromised patients, better prognosis
Secondary: due to a transformation from another type of lymphoma, worse prognosis
What characterized mature B cell lymphomas (TdT-) in children?
They are usually aggressive (either large cell, Burkitt’s, or high-grade follicular)
What morphology is associated with Diffuse Large B-cell Lymphoma?
Large, transformed cells greater than 2x small lymphocyte size – the only important cytological feature is the plasmablastic type: found in oral cavity, associated with HIV/EBV, very aggressive
What two diseases should be considered in a patient with an extremely large spleen?
Chronic Idiopathic Myelofibrosis and Hairy Cell Leukemia
What are the common sites of involvement of Lymphoplasmacytic Lymphoma?
Lymph nodes (B-cells); Marrow (plasma cells); Blood (Rouleaux formation); Spleen;
rarely extranodal
What is Waldenström Macroglobulinemia and what is it often associated with?
Proliferation of lymphocytes/plasma cells accompanied by a monoclonal IgM spike leading to hyperviscosity of the blood – often associated with Lymphoplasmacytic Lymphoma (although it can also occur with non-Hodgkin lymphoma, rarely)
How does the treatment of Hairy Cell Leukemia differ from that of other lymphomas?
It requires a special treatment consisting of purine nucleoside analogs
What sets Hairy Cell Leukemia apart from Mantle Cell Leukemia?
It does not have the t(11;14) or BCL-1 rearrangement
What immunophenotype is associated with Hairy Cell Leukemia?
Positive for CD103, CD22, CD25, CD11c, Cyclin-D1 (like Mantle), CD19, CD20, TRAP stain;
Negative for CD5, CD10, CD23
In which lymphoma is the red pulp of the spleen affected instead of the white pulp?
Hairy Cell Leukemia – also presents with “hairy” lymphocytes, marrow involvement, but rarely lymph node involvement
What is the key genetic factor for Burkitt’s lymphoma?
t(8;14) involving c-MYC)
What is different about the relapse rate for Burkitt’s lymphoma vs. other lymphomas?
Relapse usually occurs very quickly; if the patient stays disease free for 2 years, they are considered cured (vs. 5 years for other lymphomas
What lymphoma presents witth pancytopenia and monocytopenia?
Hairy cell leukemia – monocytes are unable to compensate for the neutropenia because the lymphocytes secrete an inhibitory molecule to the monocytes
What is the most common lymphoid tumor in African Americans?
Plasma Cell Myeloma (second most common in Caucasians)
What factors affect the prognosis for Plasma Cell Myeloma?
Renal insufficiency means worse prognosis; Smoldering or indolent variant means better prognosis
What immunophenotype is associated with Plasma Cell Myeloma?
Positive for CD38, CD138; Negative for surface Ig, CD20 (Rituximab not effective)
What is the most common Ig made by plasma cells in Plasma Cell Myeloma?
IgG – followed by IgA (others are rare)
What is needed to make a diagnosis of Plasma Cell Myeloma?
Clinical, Pathologic, and Radiographic diagnosis – for the initial diagnosis, order both an SPEP and a UPEP so that diseased patients aren’t missed
What is meant by the two-hit hypothesis of Plasma Cell Myeloma?
People require both an antigenic stimulus and a mutagenic event to develop the disease
What immunophenotype is associated with Lymphoplasmacytic Lymphoma?
Positive for surface IgM, CD19, CD20, CD22; Negative for CD5, CD10, CD23
What is the prognosis for Lymphoplasmacytic Lymphoma?
Indolent, incurable; worse prognosis with advanced age and cytopenias; can transform to large cell lymphoma
What clinical findings are associated with Plasma Cell Myelomas?
Osteolytic lesions in flat bones (visible on x-ray); Bone pain; Hypercalcemia; Infections (due to decreased normal antibody production); Renal insufficiency/failure (damage to renal tubules); Anemia (marrow is packed with plasma cells)
What are the cutaneous T-cell lymphomas?
Mycosis fungoides/Sézary syndrome, Primary cutaneous anaplastic large cell lymphoma, Lymphomatoid papulosis
What is the classical morphology associated with MF?
T-cells with cerebriform nuclei (cerebriform cells); Epidermotropic T-cell infiltrates; Pautrier microabscesses (highly characteristic, but rare)
What is the most common skin T-cell lymphoma and what characterizes it?
Mycosis Fungoides (MF) – characterized by scaly skin eruptions (patches and plaques), which progress to generalized plaques over the years, which then become tumors; indolent course
What cytokine-related symptoms can be seen at diagnosis with T-cell lymphomas?
Hypercalcemia (due to osteoclast activating factor); Hemophagocytic syndromes
How can T cell lymphomas be differentiated on a molecular level?
Whether they are TdT+ or TdT-; TdT+ lymphomas are precursor or thymic T-cell derived lymphomas whereas TdT- lymphomas are peripheral or post-thymic derived lymphomas
What distinguishes B cell and T cell lymphomas morphologically?
T cell lymphomas are highly polymorphic whereas B cell lymphomas are not
What are the extranodal based T-cell lymphomas?
NK/T-cell lymphoma (nasal type), Enteropathy-type T-cell lymphoma, Hepatosplenic T-cell lymphoma, Subcutaneous panniculitis-like T-cell lymphoma
How are T-cell lymphomas categorized?
By their clinical presentation: leukemic/disseminated, nodal, cutaneous, extranodal
What are the leukemic/disseminated T-cell lymphomas?
All of the ones ending on leukemia: T-cell prolymphocytic leukemia, Large granular lymphocytic leukemia, Aggressive NK cell leukemia, Adult T-cell leukemia/lymphoma
What are the nodal based T-cell lymphomas?
Angioimmunoblastic T-cell lymphoma, Peripheral T-cell lymphoma, Anaplastic large cell lymphoma
Which factors indicated a worse prognosis for a patient with MF?
>60 years of age; Increased LDH; Large T-cell transformation
How is a key aspect of ATLL diagnosis?
Clonal HTLV-1 integration – (immunophenotype is similar to that of MF)
What are the four clinical variants of ATLL and what characterizes each?
Acute (survival 2wks-yr): most common, constitutional symptoms, leukemic phase with eosinophils, skin rash/generalized adenopathy/hepatosplenomegaly, hypercalcemia (can be lethal); Lymphomatous (2wks-yr): adenopathy without leukemic phase, hypercalcemia less common;
Chronic (>2yrs): skin lesions, no hypercalcemia, increased WBC, 25% of cases progress to acute;
Smoldering (>2yrs): normal WBC, skin/lung disease, no hypercalcemia, 25% of cases progress
What is characteristic of the morphology of ATLL and what are the sites of involvement?
High nuclear:cytoplasmic ratio lymphocytes; Flower cells (or cloverleaf cells=polylobated nuclei); Sites of involvement: lymph nodes, spleen, blood, marrow, extranodal (50%: commonly skin with rare Pautrier-like microabscesses)
Which T-cell lymphoma is associated with Human T-cell Leukemia Virus-1 (HTLV-1) and what characterizes this lymphoma?
Adult T-cell leukemia/lymphoma (ATLL) – has a long latency and requires a “second hit”; mostly endemic to Japan, Caribbean, and Central Africa
What clinical signs are seen with Sézary syndrome?
Erythroderma, lymphadenopathy, circulating tumor cells, pruritus, alopecia, palmar/plantar hyperkeratoses
What distinguishes Sézary syndrome from MF?
Sézary syndrome will have >1000 MF-type cells/μL – morphologically, Sézary syndrome can also have Lutzner cells (small) and classical Sézary cells (large); there is very little bone marrow involvement; immunophenotype is similar to MF, but there is an increased CD4:CD8 blood ratio
What is the clinical staging of MF?
Stage I: confined to skin; Stage II: enlarged lymph nodes (but negative!); Stage III: histologically positive lymph nodes; Stage IV: visceral dissemination
What are the histologic grades of lymph node involvement?
Grade I (no involvement): dermatopathic, cerebriform cells; Grade II (early involvement): partial architectural effacement; Grade III (extensive involvement): complete architectural effacement
What immunophenotype is associated with MF?
Positive for CD2, 3, 4, 5, HECA; Negative for CD7, 8, ALK – MF is of peripheral epidermotropic T-cell origin
What characterizes the morphology of Anaplastic large cell lymphoma?
Hallmark cells: large cell, kidney bean/horseshoe nucleus, eosinophilic Hoff region, abundant cytoplasm, concentrate around vessels
What immunophenotype is associated with Peripheral T-cell lymphoma, NOS?
Not very specific, but CD4+>>CD8+ – Peripheral T-cell in origin
What is characteristic of the morphology of Peripheral T-cell lymphoma, NOS?
Cells with clear cytoplasm (!); Polymorphous; Inflammatory background; Hemophagocytosis
What is the most common T-cell lymphoma and how does it present?
Peripheral T-cell lymphoma, NOS: presents with eosinophilia, pruritus, hemophagocytic syndromes, lymph node and extranodal tissue involvement – aggressive but potentially curable
How does ALK affect the prognosis of Anaplastic large cell lymphoma?
The prognosis is worse if cells are ALK negative
How do cells in Anaplastic large cell lymphoma become ALK positive?
Due to t(2;5) where the ALK gene is translocated beside the Nucleophosmin gene; since Nucleophosmin is transported to the nucleus, ALK will also be found in the nucleus, thus causing it to stain positive – ALK gene codes for a tyrosine kinase receptor of the insulin superfamily
What immunophenotype is associated with Anaplastic large cell lymphoma?
Positive for CD2, CD4, CD30(!), ALK-1(!); Negative for CD3, 5, 7, 8, EBV
What is the cell of origin of ATLL and what is the usual cause of death with this disease?
CD4+ peripheral T-cell – Cause of death is usually infectious or hypercalcemia
Which T-cell lymphoma is ALK-protein positive and what characterizes it?
Anaplastic large cell lymphoma: generally a childhood disease, presents with lymphadenopathy and marrow disease, extranodal infiltrates are common (e.g. skin)
What are the two types of Anaplastic large cell lymphoma?
Primary systemic type: involves lymph nodes, marrow, and extranodal sites; Primary cutaneous type: involves skin
What are characteristic features of Hodgkin lymphoma?
Reed-Sternberg (RS) cells with possible T-cell rosettes; Non-neoplastic inflammatory background
What immunophenotype is associated with NLPHL?
L&H cell B-cell immunophenotype (i.e. doesn’t stain like a RS cell): positive for CD45, CD20, BCL-6 and negative for CD15, CD30; Also, rosettes of CD3-positive T-cells are seen around the L&H cells
What describes the lymph nodes in NLPHL?
Total to partial effacement with nodular to nodular-diffuse pattern
What is the RS cell variant seen in NLPHL?
L&H cells (aka popcorn cells); they are seen in a background of non-neoplastic lymphocytes
What clinical features are associated with NLPHL?
5% of Hodgkin lymphoma; Predominantly males, 30-50 years; Not associated with EBV; Frequent relapses, but rarely fatal; Sites of involvement are commonly the lymph nodes (extranodal is rare)
What sets Nodular Lymphocyte Predominant Hodgkin Lymphoma (NLPHL) apart from the classical type?
Although there are RS cells present, NLPHL has a non-Hodgkin phenotype
What clinical features are commonly found with Hodgkin lymphoma?
Lymph node in origin (often cervical nodes); Localized disease at presentation; Seen in younger adults; Generally excellent outcomes
What is particular about the incidence of Hodgkin lymphoma and how does this differ from other lymphomas?
The incidence is stable whereas with other lymphomas the incidence is rising (due to the increased number of immunocompromised patients)
What do all Hodgkin lymphomas have in common?
Reed-Sternberg cells (present in nodular lymphocyte predominant and classical types)
What is a key difference between Hodgkin and non-Hodgkin lymphoma?
In non-Hodgkin lymphoma malignant cells are in the majority (may have rare Reed-Sternberg cells); In Hodgkin lymphoma there are just a few malignant cells (Reed-Sternberg cells)
What is the age distribution of Classical Hodgkin lymphomas?
Bimodal: 15-35 years and late in life
What causes almost all Classical Hodgkin lymphomas in developing countries and immunosuppressed patients?
Latent Type II EBV infection
What cytokines/chemokines are associated with Classical Hodkin lymphomas?
Interleukins; GM-CSF, Lymphotoxin-a; TGF-β (leads to broad bands of fibrosis); Eotaxin (leads to the classic background of eosinophils)
What stage disease does Classical Hodgkin lymphoma usually present as?
Stage I/II: localized with a large mass; at higher stages, when the bone marrow becomes involved, large nodules (dark purple) may be seen in the marrow
What are common sites of involvement with Classical Hodgkin lymphomas?
Non-axial lymph nodes: cervical, mediastinal, axillary – a lot of patients will have a classic mediastinal mass
What are the morphologic findings of Classical Hodgkin lymphomas?
Multinucleated RS cells, mononuclear Hodgkin cells (variants), mummified cells (large pyknotic cells), T-cell rosettes, collagenous fibrosis
What clinical signs are associated with Classical Hodgkin lymphomas?
Peripheral lymphadenopathy (1-2 nodal regions); B symptoms (40%); Mononucleosis is associated with an increased risk
What is the survival associated with NLPHL?
Stage I/II: 80% survival; Stage III/IV: unfavorable; 3-5% of cases transform to Large Cell Lymphoma
What immunophenotype links together the Classical Hodgkin lymphomas?
Positive for CD15, CD30, and negative for CD45 (although they differ in clinical features, RS cell features, cellular background, and EBV rate)
What are the genetics associated with NLPHL?
There is a high rate of on-going VDJ somatic mutations; cell of origin is the germinal center B-cell (centroblastic stage) – same goes for Classical Hodgkin lymphomas
What are the clinical findings associated with Mixed Cellularity Classical Hodgkin lymphoma?
2nd most common subtype; Adult males, no bimodal distribution; Stage III/IV disease; B symptoms common; High EBV rate (75%) so more prevalent in HIV and developing countries
What describes the cellular background of Lymphocyte Depleted Classical Hodgkin lymphoma?
It is very hypocellular although RS cells are still present
What characterizes Lymphocyte Depleted Classical Hodgkin lymphoma?
Rare; Adult males; Stage III/IV disease; 80% B symptoms; EBV rate > 95% so seen in HIV and developing countries
What are the sites of involvement and growth pattern of Lymphocyte Rich Classical Hodgkin lymphoma?
Sites of involvement: mainly lymph nodes; Growth pattern: nodular (commonly) or diffuse;
Cellular background consists predominantly of small lymphocytes with few or absent eosinophils and PMNs
What characterizes Lymphocyte Rich Classical Hodgkin lymphoma?
Rare; 60 years median age; Mostly males; Stage I/II disease; B symptoms are rare; Similar clinically to NLPHL
What is an unusual characteristic of the cellular background associated with Mixed Cellularity Classical Hodgkin lymphoma?
It can present with histiocytes/macrophages and possible granuloma-like clusters (don’t confuse with cat scratch disease)
How do sites of involvement and growth pattern differ between the Mixed Cellularity type and the Nodular Sclerosis type?
In Mixed Cellularity lymph node involvement is common but it rarely is a mediastinal disease; The growth pattern in Mixed Cellularity is diffuse and not nodular as seen with Nodular Sclerosis
Which Classical Hodgkin lymphoma subtype is associated with Lacunar variant RS cells and what characterizes this disease?
Nodular Sclerosis Classical Hodgkin lymphoma: 70% of cases, young adult, 40% B symptoms, 10-40% EBV rate
What are common sites of involvement with Nodular Sclerosis Classical Hodgkin lymphoma and what distinguishes the growth pattern?
Sites of involvement: mediastinal disease, bulky disease, spleen/lung; Growth pattern: forms collagen bands around nodules (thus it is called the nodular sclerosis type)
What are the goals in the various phases of drug development?
Phase 1: pharmacokinetics (measured in all phases) and dose-depend safety; Phase 2 (open label): safety and therapeutic benefit (efficacy); Phase 3 (blinded controls, larger population): safety and efficacy; Phase 4: post-marketing surveillance
Which resources at WVU assist in setting up clinical trails?
Clinical Trials Research Unit (MBRCC) and Office of Research Compliance
What is CITI training?
Collaborative Institute Training Initiative – required for all WVU investigators engaged in human research
What approval is required for human drug development clinical trials on a local level and what does it consist of?
IRB approval (committee decision); they look at the investigators (expertise, CITI training, conflict of interest), background (what’s known about the drug), experimental design, risk/benefit (checked by Data Safety Monitoring Board), consent form (written at 6th grade level; risks, potential side effects, voluntary participation/withdrawal, alternatives, fate of data, compensation for participants), HIPPA
What are the three key principles regarding US Government guidelines for human subject research?
Beneficence (benefit/risk); Justice (equitable subject selection); Respect for persons (informed consent)
What is listed in the US Pharmacopedia Drug Information (USP DI) for Health Care Professionals?
Accepted and unaccepted off-label uses for FDA-approved drugs
What is the only step needed to be performed for generic drug development?
Companies have to prove pharmaceutical and bioequivalence to the innovator’s drug (i.e. pharmacokinetics for oral solids)
What does it mean when a cell stains positive for non-specific esterase (NSE)?
It is a monocyte
What characterizes AML, M4 (myelomonocytic)?
1/4 of AML, intermediate disease; Marrow failure; >20% blasts; >20% PMNs; >20% Monocytic; Cell of origin: PMN-monocyte split precursor; MPO-SSB/NSE positive; Immunophenotype: CD13, CD33, CD34 (for myeloblasts), CD14, CD4, CD11b/c, CD36 (for monoblasts)
What category is M3?
Promyelocytic leukemia (has
been classified)
What characterizes AML, M2 (myeloid with maturation)?
1/3 of AML, intermediate disease; Marrow failure; >20% blasts; >10% maturing neutrophils; <20% monocytes; Neutrophil dysplasia; MPO-SSB positive/NSE negative; positive for CD13, CD33, CD34, CD15 (more mature), cMPO
What characterizes AML, M1 (myeloid without maturation)?
Not as rare, aggressive disease; Marrow failure; >20% blasts; <10% maturing neutrophils; MPO-SSB positive/NSE negative; Immunophenotype: positive for CD13, CD33, CD34, cMPO
What characterizes AML, M0 (myeloid with minimal differentiation)?
Rare, poor prognosis; Marrow failure; >20% blasts, no Auer rods; Myeloid in origin; MPO-SSB/NSE negative; Immunophenotype: positive for CD13, CD33, CD34
What does it mean when a cell stains positive for periodic acid-Shiff (PAS)?
It is an RBC or a megakaryocyte
What is the difference between myelodysplasia and acute myeloid leukemia (AML)?
AML has greater than 20% blasts in blood or marrow (unless there are specific cytogenetic changes – e.g. t(8;21), t(15;17), inv(16))
What does it mean when a cell stains positive for myeloperoxidase (MPO-SSB) or Sudan Black B (SBB)?
It is a neutrophil
What does the presence of an Auer Rod indicate?
A myeloid malignancy (AML)
What characterizes AML, M5a (Monoblastic) and M5b (Monocytic)?
M5a (young people), M5b (adults); Not very common; Swelling of gingiva, bleeding; >20% blasts; >20% monocytic; <20% PMNs; no Auer rods; MPO-SSB negative/NSE positive; Immunophenotype: positive for CD13, CD33, CD14, CD4, CD11b/c, CD36
What characterizes AML, M7 (Megakaryoblastic)?
Rare, all ages; Associated with mediastinal germ cell tumors (males) and Down syndrome; Presents with bleeding gums and skin involvement; >20% megakaryoblasts; Myelofibrosis (dry tap); PAS-positive/NSE-punctate/MPO-SSB-negative; Immunophenotype: positive for CD13, CD33, CD41, cCD61
What characterizes AML, M6b (Pure Erythroid)?
Very rare, very poor outcome, all ages; >80% erythroid cells; No myeloblast increase; PAS-positive, MPO-SSB/NSE negative; Immunophenotype: positive for Glycophorin A, Heme A –
Must consider B12/folate deficiency!
What characterizes AML, M6a (Erythroleukemia)?
Rare, poor outcome, adults; Anemia with normoblastemia; >50% erythroid cells of all maturing stages; >20% of non-erythroid cells are blasts; PAS positive, Iron ± ringed sideroblasts, MPO-SSB-positive myeloblasts, NSE negative; Immunophenotype: positive for CD13, CD33, Glycophorin A, Heme A;
What characterizes AML, inv(16) with CBF-beta/MYH11?
More common, usually younger people; M4 morphology, but blasts can be <20%; Auer rods may be present, atypical eosinophils, increased monocytes, decreased mature PMNs; MPO-SSB positive/NSE weak positive; Immunophenotype: positive for CD13, CD33, cMPO, CD14, CD4, CD11b/c, CD64 – good response with Cytarabine
What characterizes Precursor T Lymphoblastic Leukemia/Lymphoma?
Young adults; 10% have leukemia/80-85% have lymphoma; Mediastinal mass is common;
Genetic translocations are common (TCR genes); MPO-SSB negative/NSE punctate; Immunophenotype: positive for TdT, CD1a/2/3/4/5/7/8; Prognosis similar to B cell ALL
What is the prognosis for Precursor B Lymphoblastic Leukemia/Lymphoma?
Pediatric: good cure rate; Adult: not as good; Prognostic parameters that are associated with a good prognosis are hyperdiploid, young age, low-normal WBC, initial response to therapy
What characterizes Precursor B Lymphoblastic Leukemia/Lymphoma?
Children; 80-85% have leukemia/10% have lymphoma; Mediastinal mass is rarely seen;
MPO-SSB negative/NSE punctate; Immunophenotype: positive for TdT, HLA-DR, CD19, CD10, CD22; Genetics (e.g. hypo- vs. hyperdiploidity) are important in prognosis
What percent of acute leukemias are myeloid and what are the associated clinical pictures of AML and ALL?
70% are AML; AML: adults (mean=60), variable Auer rods, CD13, CD33, CD34, MPO, NSE;
ALL: children (under 6), TdT positive, CD19/22 (B cell), cCD3/CD2 (T cell)
What are causes of therapy-related AMLs?
Alkylating agents/radiation therapy (e.g. for breast cancer): starts as MDS with subset going to AML, Multidrug Resistance Glycoprotein (MDR-1) presence common;
Topoisomerase II inhibitor therapy (e.g. with Etoposide treatment): presents as AML without prior MDS, usually monocytic in origin (M4/M5), cytogenetics can be 11q23 and other translocations
What characterizes AML, 11q23 with MLL translocation?
Less common, infants/children; Can be therapy-related; DIC common; Often involvement of skin/gingiva (i.e. monocytic); Involves MLL 11q23 and one of many other genes; M5 morphology (sometimes M4), monocytic with monoblasts/promonoblasts; MPO-SSB negative/NSE positive; Immunophenotype: positive for CD13, CD33, CD14, CD4, CD11b/c, CD36
What translocations are associated with t(8;21), t(15;17), inv(16), and 11q23?
t(8;21): M2 with AML/ETO1; t(15;17): M3 with PML/RARα;
inv(16): M4eo with CBF-beta/MYH11; 11q23: M5 with MLL
What characterizes AML, M3 (Acute Promyelocytic Leukemia)?
t(15; 17) with PML/RARα (retinoic acid receptor); Any age; DIC common; Lost of Auer rods (bundles of kindling – Faggot cell); >20% blasts + promyelocytes; MPO-SSB strong positive/NSE weak positive; Immunophenotype: positive for CD33, cMPO, negative for CD34 (since there are promyelocytes, not blasts); Treatment with all trans-retinoic acid (differentiating agent) and subsequent anthracycline (consolidation)
What characterizes AML, t(8;21) with AML1/ETO?
Most common cytogenetic change; Younger patients; M2 morphology, but blasts can be <20%; Variable PMN dysplasia with Auer rods, increased eosinophils; MPO-SSB positive/NSE negative; Immunophenotype: positive for CD13, CD33, CD34, cMPO – good response with Cytarabine
What is a key difference between the treatment of AML and ALL?
In AML a maintenance regimen is not normally given
What cytogenetics will be associated with about 30% of adult ALL patients?
t(9;22) – 190kd BCR/Abl
What cytogenetics are associated with Acute Promyelocytic Leukemia?
t(15;17)
What are B symptoms?
Fever, nightsweats, weight loss ( >10%/6 months) – they imply a worse prognosis
What are the cytogenetics associated with AML prognosis?
Good prognosis: t(8;21), t(15;17), inv16; Poor prognosis: -5, -7, trisomy 8, complex karyotype, prior MDS ; Intermediate prognosis: normal karyotype (largest subgroup)
What finding is diagnostic of acute myeloid leukemia?
Auer rod in cytoplasm
What is the major determinant for long-term disease survival in adults with AML?
Cytogenetics
How does age of onset differ between AML and ALL?
AML: mostly an adult disease (40% long-term survival); ALL: mostly a childhood disease
What is an abnormal feature of a bone marrow slide?
Homogeneity
What is a common site for a bone marrow biopsy?
Posterior iliac crest
What age group is mostly associated with CML?
Young adults
What stages are associated with lymphoma?
Stage 1&2: involved lymph nodes on one side of diaphragm; Stage 3: involved lymph nodes on both sides of diaphragm; Stage 4: marrow involvement
What percent of patients with Multiple Myeloma will relapse after treatment?
~95% (it is considered incurable)
In what disease are “punched-out” lytic lesions of the skeletal system found?
Multiple myeloma (due to local accumulations of plasma cells)
What mutation is thought responsible for the overgrowth of myeloid cells in Polycythemia Vera, Essential Thrombocytosis, and Myelofibrosis?
A mutation in just another kinase (JAK2) which causes it to become constitutively activated
What are the three phases of CML?
Chronic: minimal blasts; Accelerated: 10-19% blasts; Blast phase: >20% peripheral blasts
What is the pathogenesis of CML?
t(9;22) – 210, 230kd BCR/Abl
What are the WHO classifications of ALL and which is most common?
Precursor B-cell ALL (most common); Precursor T-cell ALL; Burkitt (mature B-cell) ALL
What percent of patients receiving an allograft bone marrow transplant will have bacterial infections?
85-90% - patients will receive prophylactic antibiotics, antifungals (early post-transplant), and antivirals (since Herpes simplex and CMV will reactivate – CMV can come from donor or host)
What are the chances of finding a sibling HLA matched donor?
25%
What are the most important HLA subclasses that need to be typed?
A, B, C, DR (chromosome 6); done through gene sequencing – HLA-C is not important in cord blood transplants
What are the two most common reasons to go through an autologous bone marrow transplant?
Multiple Myeloma and Hodgkin’s Lymphoma – autologous = patient’s own stem cells rescue immune system after chemotherapy
How does therapy differ for patients over the age of 50?
They do not receive ablative therapy or radiation, but rather get reduced intensity immunosuppressive treatments (non-ablative)
What is the difference between bone marrow, peripheral blood, and cord blood grafts regarding allografts?
Bone marrow: mid stem-cell/T-cell dose, 20-25 days for engraftment, mid chances of GVHD; Peripheral blood: high stem-cell/T-cell dose, 10-14 days for engraftment, mid chances of GVHD; Cord blood: low stem-cell/T-cell dose, 30-40 days for engraftment, low chances of GVHD
What are the most common reasons to have an allogeneic bone marrow transplant?
Leukemia, aplastic anemia, refractory lymphoma – patients have abnormal bone marrow
Reactivation of what virus is seen in almost all autologous transplant patients?
Varicella zoster – prophylactic Acyclovir
What types of infections are common in the first 14 days after transplant?
Gram positive/negative, fungal (Candida), Herpes simplex
What happens to the innate and acquired immune systems after an autologous transplant?
The innate comes back quickly, whereas the acquired immune system takes 6-12 months to recover
What organs are affected most in Graft vs. Host disease?
Skin, GI tract, liver – about 50% of patients will be affected by GVHD
What is the key difference between Graft vs. Tumor effect and GVHD?
Graft vs. Tumor occurs through direct cell contact, whereas GVHD occurs via an APC
What is the most common teratoma of childhood?
Sacrococcygeal teratomas: girls>boys, 75% benign/10% malignant, mostly seen in younger infants
What morphology is associated with neuroblastoma?
Wide size range; Soft, gray, brain-like tissue: may show necrosis, cystic softening, hemorrhage and calcification; Small primitive cells (neuroblasts): can be seen as Homer Wright pseudorosettes or ganglion cell differentiation
What are neuroblastic tumors derived from and where do they occur most frequently?
Derived from primordial neural crest cells; Occur mostly in the abdomen (adrenal medulla and sympathetic ganglia) but also in other sites like the posterior mediastinum
What genetic markers are associated with small, round, blue cell tumors of childhood?
Ewing sarcoma/PNET: CD99 expression; Rhabdomyosarcoma: Myo D1 expression; Burkitt lymphoma: t(8;14); Lymphoblastic lymphoma: TdT+; Wilms tumor: chromosome 11 (WT1 gene); Retinoblastoma: 13q14 (RB) deletion/mutation
What is the differential for a small, round, blue cell tumor?
Neuroblastoma, Lymphoma, Rhabdomyosarcoma, Ewing sarcoma/PNET
What malignancy is associated with Down Syndrome?
Leukemia
What is the leading cause of death from disease in children 4-14 years of age?
Malignant neoplasia (top three: leukemia, CNS tumors, lymphoma)
What are the two peaks of incidence associated with teratomas?
Within first 2 years: congenital; Late adolescence: slow growing
What are the benign pediatric tumors?
Hemangioma, Lymphangioma, Fibrous tumors, Teratomas
How does histologic maturity of teratomas relate to its biologic behavior?
Mature: benign, well-differentiated cystic lesions; Immature: lesion of indeterminate malignant potential; Malignant: mixed with another germ cell tumor component
What are the stages of neuroblastoma?
1: localized tumor;
2A: localized with incomplete gross excision, ipsilateral lymph nodes negative;
2B: : localized with incomplete gross excision, ipsilateral lymph nodes positive;
3: unresectable unilateral tumor infiltrating across the midline:
4: metastasis to distant lymph nodes
What do the letters in WAGR syndrome stand for?
Wilms’ tumor; Aniridia; Genital anomalies (e.g. hypospadias); mental Retardation
What is the function of the WT1 gene?
Encodes a transcription factor expressed in the kidneys and gonads of the developing fetus – found on chromosome 11
What syndromes/malformations are associated with an increased risk for Wilms’ tumor?
WAGR syndrome, Denys-Drash syndrome, Beckwith-Wiedemann syndrome
What is the most common primary renal tumor of childhood and what age of diagnosis is associated with it?
Wilms’ tumor (nephroblastoma); diagnosed between ages 2-5
How does DNA ploidy affect the prognosis of neuroblastoma?
Diploid is unfavorable (this is the opposite for most diseases)
What is meant by stage 4S regarding neuroblastoma?
Localized primary tumor (1, 2A, 2B) with dissemination limited to skin/liver/bone marrow – limited to infants < 1year of age
What is the clinical presentation associated with neuroblastoma?
<2 years of age: large abdominal mass, fever, possible weight loss; >2 years of age: possible metastatic disease, bone pain, respiratory symptoms, GI complaints – neonates can present as “blueberry muffin babies” (stage 4S)
How does age of the child affect prognosis of neuroblastoma?
< 1 year: excellent prognosis; 1-5 years: intermediate prognosis (depending on tumor stage); >5 years: very poor prognosis
What biologic markers are associated with neuroblastoma?
Catabolites of catecholamines: VMA, HVA; Serum neuron specific enolase; N-myc amplification
What syndrome is associated with a gene on 11p15.5 (“WT2 gene”) and what are the clinical findings?
Beckwith-Wiedemann syndrome: enlargement of body organs, macroglossia, hemihypertrophy, omphalocele, adrenal cytomegaly
What is the prognosis for Wilms’ tumors?
With chemotherapy, radiotherapy, and surgery, survival is 90% - diffuse anaplasia is considered unfavorable (50% 2-year survival)
What does anaplasia in a Wilms’ tumor correlate with?
Underlying p53 mutations and resistance to chemotherapy – found in 5% of Wilms’ tumors
What is the morphology of a Wilms’ tumor?
Triphasic pattern containing blastema, stroma, and epithelial cells
What is the clinical presentation of Wilms’ tumor?
Large abdominal mass; also hematuria, pain in abdomen, intestinal obstruction, hypertension, pulmonary metastasis
What does the presence of nephroblastomatosis imply an increased risk of?
Wilms’ tumor (nephroblastomatosis refers to the presence of a normal tissue at an abnormal time; also known as nephrogenic rests)
What tumors are children with Beckwith-Wiedemann at increased risk for?
Wilms’ tumor, hepatoblastoma, adrenocortical tumors, rhabdomyosarcomas, pancreatic tumors
What gene, found in the same chromosomal region as WT1, is associated with aniridia?
PAX6 (both are located on 11p13) – if a patient has a deletion restricted to PAX6, they will not be at increased risk for a Wilms’ tumor
What clinical findings are found with Denys-Drash syndrome?
Gonadal dysgenesis and nephropathy – due to germline abnormalities of the WT1 gene (dominant negative missense mutation)
Which bacteria, normally found in the gut, are a common cause of UTIs?
E. coli
What is the classical explanation of why certain cells stain Gram+ and others Gram-?
Gram+ cells have a very thick cell wall
What cell arrangements are often seen with Streptococcus and Staphylococcus, respectively?
Streptococcus: chain-like; Staphylococcus: grape-like
What terms describe the shapes of bacteria and what are examples associated with each?
Cocci (speherical): Streptococcus, Staphylococcus; Bacillus (rods): Bacillus anthracis, E. coli;
Vibrio (curved): Vibrio cholerae; Spirochete (spiral): Treponema pallidum, Borrelia burgdorferi
Why is the difference between eukaryotic and prokaryotic ribosomes important?
Prokaryotes have 70S ribosome (vs. 80S in eukaryotes); the 70S ribosome differs in DNA sequence which makes it vulnerable to attack by Gentamycin and Neomycin
How do Mycoplasma bacteria differ from other bacteria?
They do not have peptidoglycan, but do have sterols in their cell membrane
Which bacteria, normally found in the gut, can cause “gas gangrene” in wounds?
Clostridium perfringens
What are the main classifications of microbes based on?
Cellular with nuclear membrane: parasites, fungi, yeasts; Cellular without nuclear membrane: bacteria; Not cellular: viruses, prions
What are some examples of microbes that are commensal and how do they help?
E. coli: vitamin K; various bacteria: vitamin B12; lactobacilli: acidify vagina and prevent Candida albicans overgrowth – most importantly, commensal bacteria compete with pathogenic species for colonization
What size range is associated with microbes?
Bacteria: 0.2 μm (Mycoplasma) – 700 μm (Thiomargarita);
Viruses: 20 nm (polio) – 200 nm (smallpox)
What is a key difference in the peptidoglycan between Gram+ and Gram- bacteria?
Gram+ bacteria use (Gly)5 between the two peptides forming the cross-link; Gram- bacteria use a L-DAP instead of a L-lysine connecting the D-ala forming the isopeptide bond
Which antibiotics block synthesis of the tetrapeptide precursor of cell wall synthesis?
Fosfomycin (for UTIs), cycloserine (for TB)
What is the mechanism of action of Bacitracin?
It blocks peptidoglycan precursor synthesis by blocking regeneration of the lipid carrier (BP)
What is the mechanism of action of Vancomycin?
It blocks the glycosyltransferase (blocks chain polymerization); it does so by binding to the terminal D-ala/D-ala structure
What is the mechanism of action of Penicillin?
As a structural analog of the D-ala/D-ala dimer, it inhibits the transpeptidase reaction by forming a covalent complex with them
What external reactions are involved in the biosynthesis of cell wall?
Glycosyl transfer (polymerization of NAG/NAM structures) --> transpeptidase cross-linking --> carboxypeptidase removes a terminal D-ala on one of the peptides
What cytoplasmic reactions are involved in the biosynthesis of cell wall?
NAG --> NAM --> NAM with L-ala/D-glu/L-DAP --> NAM with L-ala/D-glu/L-DAP/D-ala/D-ala --> switch terminal UDP for bactoprenol (BP) --> attach NAG --> attach (Gly)5 in Gram+ --> flip entire structure to outside of cell wall
What is the major component of bacterial cell walls and what does it consist of?
Peptidoglycan: composed of alternating glycans (NAG/NAM) with attached peptides (D-ala, L-DAP, D-glu, L-ala)
How does lysozyme affect peptidoglycan?
It cleaves the β1-4 linkage between NAG and NAM
Between which peptides does cross-linking take place?
D-ala / L-DAP; forms an isopeptide bond utilizing a COOH or NH2 from the R group instead of one from the normal backbone structure; cross-link occurs between tetrapepide and pentapeptide
What additional cell wall structure is found in Gram-, but not in Gram+ bacteria?
LPS (is attached to the peptidoglycan via periplasm and an outer membrane, which are not found in Gram+ bacteria)
What is the key component in LPS that causes it to be an endotoxin?
Lipid A (binds TLR-4 causing fever, inflammation, shock)
What are the components of Lipopolysaccharide (LPS)?
Conserved portion: Lipid A connected to a short polysaccharide part called the core;
Highly variable portion: long polysaccharide part called the O-antigen/O-polysaccharide
What are the antitumor antibiotics?
Doxorubicin, Epirubicin, Idarubicin, Mitoxantrone, Actinomycin D, Bleomycin
What is the active metabolite of Irinotecan and how is it excreted?
Irinotecan is converted by carboxyesterases to SN-38 (100-1000x more active); SN-38 is glucoronidated and excreted in the bile; CYP3A4 also metabolizes Irinotecan, but forms inactive metabolites
What is the mechanism of action of Irinotecan?
Stabilization of a ternary complex between topoisomerase I and dsDNA, inducing single strand breaks, thus preventing cells from entering mitosis (G2 arrest)
What are the topoisomerase I inhibitors?
Irinotecan, Topotecan
What toxicities are found with Doxorubicin?
Cardiac problems: acute arrhythmias and chronic CHF (keep lifetime dose <550 mg/m2); Vesicant; Radiosensitizer; May make patient’s fluids/tissues red
What important pharmacokinetics are associated with Doxorubicin?
It has a large volume of distribution; Elimination is via bile, so bilirubin levels must be checked
What is the most common antitumor antibiotic and what is its mechanism of action?
Doxorubicin; MOA: free radical formation, topoisomerase II interaction (!), DNA intercalation
What are the mechanisms of action of 5-Fluorouracil (5-FU)?
Inhibition of thymidylate synthase (= decreased thymidine); Incorporation into RNA and DNA leading to apoptosis – some S phase specificity
What are important toxicities associated with 5-FU and what drug is used in association with 5-FU?
Toxicities (continuous infusion): mucositis, diarrhea, hand-and-foot syndrome (paresthesias, redness --> pain --> peeling --> resolution); Toxicities (daily bolus): myelosuppression; Drug interactions: Leucovorin (folinic acid) stabilizes/prolongs binding of 5-FU to thymidylate synthase
What compound causes deactivation of 5-FU?
Dihydropyrimidine dehydrogenase (DPD); 5-FU has a short half-life (min.) – DPD deficiency often results in lethal toxicity
What important toxicities are associated with Vincristine?
Neuropathies: decreased DTR --> paresthesias --> autonomic dysfunction; SIADH; Photosensitivity; NOT myelosuppression (sets it apart) – If accidentally given intrathecal, it is almost always lethal
What is characteristic of the metabolism of signal transduction inhibitors?
They are all metabolized by CYP3A4
What are the signal transduction inhibitors used in chemotherapy?
Dasatinib, Erlotinib, Imatinib, Lapatinib, Sorafenib, Sunitinib, Temsirolimus
What sets Ixabepilone apart from Paclitaxel and Docetaxel?
Although it has a similar MOA as Paclitaxel/Docetaxel, it is less susceptible to MDR/P-gp and may therefore work on vinca/taxane/anthracycline resistant cells
What important toxicities are associated with Paclitaxel and Docetaxel, respectively?
Paclitaxel: acute anaphylaxis, myelosuppression, peripheral neuropathy; Docetaxel: myelosuppression, peripheral neuropathy, fluid retention
How are Paclitaxel and Docetaxel metabolized?
By CYP3A4 and 28C (Paclitaxel only) – they are easily cleared from the CNS thus reducing the effects there; this can lead to increased relapse rates in the brain
What is the mechanism of action of Paclitaxel and Docetaxel?
Promotes microtubule assembly and then inhibits depolymerization, thus stabilizing the microtubules in their non-functional state
What important toxicities and drug interactions are associated with Irinotecan?
Myelosuppression; n/v; Diarrhea: early (treated with atropine), late (treated with loperimide) – Drug interactions: CYP3A4 inducers/inhibitors
What are the mitotic inhibitors used in chemotherapy?
Vincristine, Vinblastine, Vinorelbine, Paclitaxel, Docetaxel, Ixabepilone
What is the mechanism of action of Vincristine and what is its half-life?
Binds tubulin, thus inhibiting microtubule formation (i.e. M phase specific = mitotic arrest) – it’s half-life is long (>20 hrs)
What is the mechanism of action of Temsirolimus?
It binds to FKBP-12 and inhibits mTOR (like Sirolimus) --> this leads to growth arrest
What is the mechanism of action of Tamoxifen and what are its important toxicities?
Competitive estrogen antagonist (mostly G2 specific) – Toxicities: menopausal symptoms, thromboembolism, flare reaction, ocular/endometrial/uterine cancer
What are the estrogen receptor modulators used in chemotherapy?
Fulvestrant, Tamoxifen, Toremifene
What are the two subclassifications of anti-estrogen therapies and what characterizes each?
Receptor modulators: used pre- or post-menopausal, toxicities include blood clots / uterine cancer;
Aromatase inhibitors: used post-menopausal, toxicities include osteoporosis / fractures
What pharmacological class is Vorinostat and what is its mechanism of action?
Histone deacetylase inhibitor – MOA: inhibits HDAC1,2,3,6 which catalyze removal of acetyl groups from histones and transcription factors --> cell cycle arrest and/or apoptosis
What pharmacological class is Bortezomib and what is its mechanism of action?
It is a proteasome inhibitor – MOA: reversible inhibitor of ubiquitin proteasome pathway
What is the pharmacologic class of Arsenic Trioxide and Tretinoin
They are differentiating agents
What two signal transduction inhibitors are competitive inhibitors of ATP on the tyrosine kinase Bcr-Abl and how do they differ?
Imatinib and Dasatinib – Dasatinib has activity in Imatinib-resistant cell lines
Which signal transduction inhibitors are associated with the EGFR/ErbB-1/ HER1 tyrosine kinases and what differentiates between them?
Erlotinib and Lapatinib – Erlotinib: causes cell cycle arrest and inhibition of angiogenesis, and is associated with a rash (no rash = no efficacy); Lapatinib has a dual action on ErbB-1 and ErbB-2, making it useful in HER-2 positive breast cancer
Which signal transduction inhibitors are used for renal cell cancer and what is characteristic of them?
Sorafenib and Sunitinib – both are multi-kinase inhibitors causing inhibition of proliferation and antiangiogenesis; since they affect multiple tyrosine kinase inhibitors, they have more extensive toxicities associated with them
What is particular about the pharmacokinetics of Tamoxifen?
It is metabolized by several CYP450 enzymes (3A, 2C, 2D6); however, in order to achieve maximal efficacy, Tamoxifen must be metabolized by CYP2D6 to highly active metabolites; since ~10% of Caucasians do not have CYP2D6, this can cause problems; Inhibitors of CYP2D6 should also be avoided
What important toxicities are associated with Leuprolide?
Hot flashes, reduced libido, impotence, gynecomastia
What is the mechanism of action of Leuprolide?
`It is a GnRH agonist --> causes an initial surge of LH/FSH from the pituitary, temporarily increasing the levels of testosterone (~ 1 week) --> ultimately secretion of LH/FSH is decreased by continuous receptor stimulation – used to treat prostate cancer (also breast/ovarian cancer)
What are the androgen receptor antagonists, used in chemotherapy and for what specific cancer are they important treatment options?
Bicalutamide, Flutamide – used in prostate cancer
What is the mechanism of action of Anastrozole and what toxicities are associated with it?
MOA: blocks aromatase conversion of androgens to estrogens in peripheral tissues; Toxicities: menopausal symptoms, vaginal dryness/bleeding, GI effects, osteoporosis
What are the aromatase inhibitors used in chemotherapy?
Aminoglutethimide, Anastrozole, Letrozole, Exemestane
What is special about the outer membrane of Gram- bacteria?
It is a permeable barrier that confers resistance to antibiotics; they get their nutrients through porins found in the outer membrane
What is the importance of efflux pumps?
They pump out all sorts of substances (including antibiotics)
Why are siderophores highly diverse between bacteria?
If a bacterium were to produce a siderophore that is recognized by other bacteria, it will allow those bacteria to make use of its siderophore, in effect reducing its own iron levels
How does iron accumulation occur in bacterial cells?
Bacteria secrete siderophores which scavenge Fe3+; once it finds iron, a periplasmic binding protein brings the siderophore to the permease, where its transport across the membrane is powered by the hydrolysis of ATP (ABC transporters)
How does lactose get into bacterial cells?
The accumulation of lactose is driven by the proton-motive force
What functions are associated with the cytoplasmic membrane of bacteria?
Energy production (H+-linked ATPase, respiratory chain with cytochromes); Transport;
Secretion (e.g. of proteins); Signaling (chemoreceptors)
Which bacteria are neither Gram+ nor Gram-?
Mycoplasmas: don’t have cell walls; Mycobacteria: have thick, waxy cell walls that require a special acid-fast stain (AFB)
\What structural component of group A strep is used for adhesion to eukaryotic target cells?
M protein (“adhesin” protein)
What is a purpose of pili/fibriae found in Gram- bacteria?
They have terminal proteins that are used to attach to eukaryotic cell components
What is the purpose of capsule on Gram+ and Gram- bacteria?
Protect against phagocytosis and protect against drying out – capsule is a slime consisting of polysaccharides
What has to happen for flagella to continue in a swimming motion and not change into the “tumble”?
The concentration of chemoattractants has to continuously increase so that it can stimulate the flagella into not reversing its motion
What is the period called that occurs before exponential growth takes place?
Lag phase
What are ways of determining bacterial growth curves?
Serial dilutions followed by plating; Spectrophotometry (measures turbidity)
What is chocolate agar?
Agar that contains lysed RBCs – used to grow certain types of bacteria
What protein is present in bacteria that can clear blood agar?
Hemolysin (this is a virulence factor for bacteria)
What is the purpose of the molecular syringe (type III secretion apparatus) associated with certain bacteria?
It allows the bacteria to insert certain small molecules into the host cells it’s trying to invade
Besides adhesion to cells, what other function is associated with pili?
Conjugation/mating between bacteria: transfer of DNA
What is the difference between ccw rotation and cw rotation of the flagellar bundle?
ccw: smooth swimming; cw: “tumble” (reorients itself)
What is an important purpose of flagella?
Chemotaxis
What propels flagella?
Proton-motive force
What compounds can be used as alternate electron acceptors?
Nitrate, DMSO, fumarate
What is the best way to kill spores?
Combination of heat and water (steam, as in an autoclave)
What can certain bacteria undergo in conditions of nutrient limitation?
Sporulation (can be done by certain Gram+ bacteria: Clostridia, Bacillus)
What are examples of “rich media”?
Digests of meat, yeast cultures, milk proteins, sometimes with blood added
What is the difference between a prototroph and an auxotroph?
Prototroph: a bacterial strain that has the same nutritional requirements as the wild-type strain from which is was derived; Auxotroph: a mutant organism that requires some nutrient not required by the organism from which it was derived
What are the enzymes that convert superoxide and hydrogen peroxide, respectively?
Superoxide: superoxide dismutase (SOD); Hydrogen peroxide: catalase – these enzymes are missing in obligate anaerobes
What happens when a pair of oxygen’s takes one, two, or three electrons?
One: formation of superoxide; Two: formation of hydrogen peroxide; Three: formation of hydroxyl radical (most damaging due to its rapid reaction rate – no enzymes can detoxify this)
406. What factor increase is seen with 10 generations of exponential growth?
1,000x (e.g. a population that doubles every 20 minutes will see a 1,000x increase in 3.5 hours)
What are requirements for a bacterial growth medium?
Water, carbon (e.g. glucose), nitrogen (NH4+, or an amino acid), inorganic ions (sulfate, phosphate, K/Mg/Fe, trace metals), oxygen
What are the different categories of bacteria associated with how they handle oxygen?
Obligate aerobes: must have oxygen (Pseudomonas, Mycobacterium tuberculosis);
Obligate anaerobes: cannot have oxygen (Clostridium, Bacteroides);
Facultative anaerobes: will respire if oxygen is present, but ferment if not (E. coli, Staph. aureus)
Urban dwellers take prescribed medications more faithfully (T/F)?
F
What is the best way to end a visit with a patient?
“Is there something else you need today”?
What factor has been shown to positively influence proper adherence to medications?
Physician job satisfaction
Older people (over 60) are more likely to take their medications (T/F)?
T
The more medications, the least likely they will be taken properly (T/F)?
F
A lower educational background promotes compliance (T/F)?
F
Females are more likely to follow physicians’ prescribing advice (T/F)?
F
Why has the nomenclature changed from compliance to adherence?
Because compliance indicates that the patient should yield to a doctor’s opinion when in fact there should be a more equal doctor-patient relationship
What is the percentage of people with hypertension who regularly take their medication?
76% (has better adherence rate than most medications)
Smokers are more compliant with medications (T/F)?
F
What are some physician factors influencing adherence?
Number of patients seen per week (the more, the worse); Scheduling of follow-up appointments (the more, the better); Physician specialty (more specialized = less adherent); Number of tests ordered (the more, the better); Physician willingness to answer questions (the more, the better)
At what level does the average American read?
8th grade level (20% 5th grade or lower) – 1 in 5 American are functionally illiterate
What percent of Emergency Department patients misunderstand their diagnosis, treatment, or follow-up instructions?
78% (34% misunderstood follow-up instructions)
What are factors related to compliance with treatment for hypertension?
Other concurrent heart disease; Belief that medications are necessary for good health; Personal perceptions and social circumstances; Females are more likely to comply with “diet” changes than with “lifestyle” changes; Educations and female gender do make a difference with diet
What is a factor that has a neutral influence on patient adherence to physician recommendations?
The seriousness of the problem being treated
What is the primary site of metabolism for recombinant drugs and why?
Kidney – since they are small, the drugs can leak through the glomerulus; however, when they are reabsorbed in the nephron, they are degraded by local proteases
What is the nomenclature used for MoAb?
Common suffix is “mab”; Antibody source: u = human, o = murine; xi = chimeric
What problems are associated with MoAb use?
Cross-reactivity with normal tissues; Antigenic modulation (receptor internalization); Development of anti-mouse antibodies by patient (can only give one dose); Tumor heterogeneity in antigen expression; Tumor lacks adequate blood supply; Lack of direct toxicity; Lack of host accessory effectors
What is particular about the half-life of monoclonal antibodies (MoAb) and what is the most important organ for clearance?
Half-life is long; Liver and reticuloendothelial system are most important organs for clearance
What two drugs are interleukins and how do they differ?
Aldesleukin (IL-2): stimulates the immune system, severe toxicities associated with it (e.g. hypotension), only used as a last resort; Denileukin (fusion protein of diphtheria toxin fragments and IL-2): targets delivery of toxin to cells expressing an IL-2 receptor, used to destroy cells in some leukemias/lymphomas
What is Alpha interferon used for and what toxicities are associated with it?
Leukemias, lymphomas, melanomas, myelomas – toxicities: flu-like symptoms, n/v, weight loss
What are the biologic effects of interferons?
Antiproliferative; Immunomodulatory; Differentiation (monocytes --> macrophages); Antigen expression (increased Ig expression on lymphocytes); Oncogene expression (reduced)
What are the nonspecific immunostimulants?
BCG; Levamisole
How does the effectiveness of treatment compare when looking at biologic response modifiers, between continuous SC infusion and bolus IV?
Continuous SC is far more effective than bolus IV
What are compounds called that modify the host’s biologic response to tumor cells?
Biologic Response Modifiers
What is the mechanism of action of Trastuzumab and what are some of its toxicities?
MOA: binds to HER2 receptor leading to cell cycle arrest, and acts as a mediator of antibody-dependent cell-mediated cytotoxicity via natural killer cells and monocytes; Toxicity: hypersensitivity, CHF (especially in combination with Doxorubicin – signs may be delayed)
What is the mechanism of action of Filgrastim and Pegfilgrastim and what are some of their toxicities?
MOA: they are G-CSF and thus stimulate multiple stages of neutrophil production as well as increase the activity of individual neutrophils; Toxicity: bone pain, hyperuricemia
What are the established roles of neutrophil-related growth factors?
Prevention (not treatment) of neutropenic fever following “standard-dose” chemotherapy; Acceleration of hematopoietic recovery following “high-dose” chemotherapy; Generation of autologous peripheral blood progenitor cells; Treatment of chronic neutropenic states
What are the indications for EPO?
Anemia secondary to renal failure, malignancy, or chemotherapy
Besides causing hematopoiesis, what negative side-effects are associated with EPO?
Cancer-related angiogenesis (via VEGF), promotion of cancer cell migration; also hypertension, thrombotic events, and seizures
What is the mechanism of action of Tositumomab?
MOA: it is murine IgG conjugated with Iodine131, which binds to CD20 on B-cells, leading to complement-dependent toxicity, antibody-dependent cellular toxicity, and direct toxicity of radioactivity (to the bound cell and adjacent cells)
What is the mechanism of action of Gemtuzumab and what are some of its toxicities?
MOA: it is humanized IgG conjugated with the toxin calicheamicin, which becomes internalized after CD33 binding on leukemic cells (causes dsDNA breaks); Toxicities: myelosuppression
441. What is the mechanism of action of Bevacizumab and what are some of its toxicities?
MOA: binds to VEGF, thus inhibiting tumor-related angiogenesis; Toxicities: hypertension, proteinuria, problematic wound healing (since it inhibits angiogenesis)
What is the mechanism of action of Cetuximab and what are some of its toxicities?
MOA: binds to EGFR (ErbB-1/HER1), and in effect competitively inhibits EGF/TGF-α binding, thus blocking signaling for tumor cell growth, angiogenesis, and metastasis; Toxicities: Acneform rash (like Erlotinib), infusion reactions
What is the mechanism of action of Rituximab and what are some of its toxicities?
MOA: binds to CD20 on B-cells (CD20 regulates cell cycle), and causes B cell lysis; Toxicities: Infusion reactions, risk of acute death – Rituximab is used for B-cell non-Hodgkin’s lymphoma
What is the difference between Filgrastim and Pegfilgrastim and what drug interactions are associated with them?
Pegfilgrastim is pegylated, thus substantially increasing its half-life (since it cannot be filtered out at the glomerulus); Both drugs should not be used concomitantly with cycle-specific chemotherapy (e.g. with 5-FU) since it can worsen myelosuppression
What is the mechanism of action of Sargramostim and what are its toxicities?
MOA: since it is a GM-CSF, it has the same MOA as Filgrastim; Toxicities: also similar to Filgrastim, but includes myalgias
What is Oprelvekin and what is its mechanism of action?
IL-11; MOA: stimulates multiple stages of megakaryopoiesis and thrombopoiesis
At what point can diagnostic techniques be used to detect a cancer?
At about 109-1010 cells
Where and how does bacterial DNA replication occur?
It occurs at a site on the DNA called the OriC; initially, levels of DnaA increase and bind around OriC à this causes DnaB/DnaC to bind as well, which functions as the helicase that separates the two DNA strands – DNA replication is bidirectional!
How does expression of the lac operon occur?
Under normal conditions, a gene found upstream from the lac operon, called lacI, is transcribed and produces a protein that binds to the operator region, thus inhibiting transcription of the operon – when lactose (or a structurally similar molecule) binds with the inhibitor protein (from lacI), it will dissociate from the operator region allowing transcription of the lac operon to take place
What does the lac operon consist of?
3 genes cotranscribed into 1 mRNA: lacZ (β-galactosidase, converts lactose à glucose/galactose), lacY (lactose permease, brings lactose into the cell), lacA
What does the bacterial RNA polymerase consist of and what gives it its specificity?
Core enzyme: 2 α-units, one β, and one β`; the protein Sigma (σ) is the specificity factor that directly recognizes the promoter site
How does control of pyrimidine synthesis occur within bacteria?
The catalytic subunits that are responsible for the formation of pyrimidines are bound by regulatory subunits which respond to nucleotide binding (i.e. if the level of CTP increases, it will inactivate ATCase so that other pyrimidines can be formed instead, in order to maintain a healthy balance in pyrimidine synthesis)
How do bacteria prevent multiple initiations at OriC?
By methylation of the origin
Where is chromosome replication regulated?
At the initiation step – once initiation occurs, replication occurs at the same rate (it is constant); what differs between different bacteria is how long it takes to initiate replication (i.e. if doubling time is 20 min., that means replication is initiated every 20 min. – says nothing about the speed of DNA elongation)
What is the difference between DNA polymerase I and III?
DNA Pol. I: replaced RNA primer with DNA; DNA Pol. III: extension of DNA on leading and lagging strands
What is the purpose of negative supercoiling?
It favors processes like transcription and replication
What is the mechanism of action of DNA gyrase and which class of antibiotics uses this mechanism to our benefit?
It puts negative supercoils into DNA by causing ds-DNA breaks and subsequently reforming them in a different configuration – Fluoroquinolone antibiotics (e.g. Ciprofloxacin) trap bacterial DNA gyrase while it is bound to ds-DNA, leading to ds-breaks in the bacterial DNA, thus killing the organism
How does the level of cAMP determine whether the lac operon is transcribed?
cAMP will associate with CRP and dimerize; this complex then binds to a promoter region and subsequently recruits RNA polymerase to initiate transcription of the lac operon – cAMP levels will be low when glucose is present so that the lac operon is not transcribed
What is the purpose of translational repression?
It prevents overproduction of regulatory proteins – for example, S20 is a regulatory protein involved in the assembly of the 16S ribosome from rRNA; S20 has a high affinity for rRNA and will bind to it if present; however, if rRNA is not present, S20 will bind to its own mRNA transcript and inhibit its translation; in this fashion it prevents its own overproduction
How do rapidly dividing bacteria speed up their production of proteins?
They produce more ribosomes (individual ribosomes do not work faster)
What structures are involved in the peptidyl transferase reactions?
RNA only – there are no ribosomal proteins found near the active site
What does attenuation cause?
Conditional termination of transcription after initiation has taken place (e.g. tryptophan operon)
What is the main function of two-component regulatory systems?
Sensing small molecules outside the cell (i.e. signal molecules); when the sensor kinase component binds the signal molecule, it dimerizes and self-phosphorylates à the phosphorylated forms then phosphorylates a cytoplasmic response regulator à this response regulator then dimerizes and becomes active, which allows it to bind to DNA to activate or repress transcription
What is the purpose of a complementation test?
To see whether two different mutations affect the same gene; if the mutations are on the same gene, no growth will occur since neither organism containing the mutation is able to compensate for the other’s disability; however, if the mutations are on different genes, the organisms will be able to grow by using the non-mutated form from their respective partner
What is an example of a common mechanism for suppression of a mutation?
In the case of a nonsense mutation (creation of a stop codon), one copy of the gene used for tRNA synthesis can mutate to read the stop codon as though it were specific for the amino acid that was mutated in the first place (thus generating a functional protein)
What does the term reversion mean?
A change in phenotype from mutant (e.g. frameshift leading to a nonsense mutation in a gene required for histidine synthesis) back to wild-type
What is characteristic of the treatment for a Mycobacterium tuberculosis infection?
Combination therapy is necessary since using any of the drugs alone rapidly leads to resistance
What is the importance of the fact that DNA uses thiamine instead of uracil?
Thiamine has an extra methyl group; this allows it to be distinguished from a deaminated cytosine (which is very similar to uracil), thus reducing errors in DNA synthesis
Which factors are responsible for the low rate of mutations seen in DNA?
Base complimentarity; Proofreading function by DNA polymerase (exonuclease domain); Mismatch repair (parental DNA strand is marked by methylation)
What is the key difference between a plasmid and a chromosome?
Plasmids don’t contain any genes essential for viability
Why is the effect of a mutation in bacterial DNA never masked (i.e. always expressed)?
Prokaryotes are haploid, so changes in DNA sequence can’t be compensated for by a second copy of the chromosome (as with diploid eukaryotes)
In what situations can a mutant genotype still present with wild-type phenotype?
If the mutation is silent or a substitution is accepted; if the mutation affects an intron (not as common in prokaryotes); if the mutation is suppressed: a second mutation compensates for the first
What is the use of conditional mutations in the study of phenotype?
They are most important in studying essential functions, since dead is a phenotype (e.g. making a protein temperature-sensitive and studying its effects on phenotype at various temperatures)
How do the bacteria in the gut differ between a normal individual and one with IBD?
The person with IBD has a much greater variety of microbes present, whereas the healthy individual has only a couple of classes of bacteria present
What side effect, associated with β-lactams, is only found in Penicillins that are penicillinase resistant (e.g. Oxacillin, Nafcillin)?
Neutropenia and interstitial nephritis (!)
What adverse effects are associated with Tetracyclines?
Discoloration of teeth in children; Hepatotoxicity in pregnancy (Aminoglycosides are also class D in pregnancy!); Photosensitivity
What adverse effects are associated with Vancomycin?
Vancomycin (prevented by giving an antihistamine and slowing the IV infusion rate)
Ototoxicity (auditory >> vestibular); “Red-Man” syndrome: histamine release due to rapid IV infusion of
What important adverse effects are associated with Aminoglycosides?
Nephrotoxicity and ototoxicity (both components of the 8th nerve) – can measure plasma levels
What unique side-effect is associated with Ceftriaxone?
Biliary sludging (reversible, do not treat!)
]What does it mean when an antibiotic is cell cycle independent?
The antibiotic can kill the cell regardless of the phase it is in; this means that killing of the cells is only dependent on the amount of drug that’s available (concentration dependent), and not on how long the drug is present (time independent)
What side effects are associated with β-lactams?
Non-allergic ampicillin/amoxicillin rash; Seizures (with high CNS levels); Increased Na/K levels (since these ions are often part of the drug preparation) – however, β-lactams are safe in pregnancy
Which type of antibiotic (cell cycle dependent/independent) would best be given as a single bolus dose?
Cell cycle independent, since it is concentration dependent and will kill the cells without regard for time – cell cycle dependent drugs should be given as a continuous infusion (if possible)
What are examples of cell cycle dependent and independent antibiotics?
Dependent: β-lactams, since they are bactericidal during cell wall synthesis; Independent: Aminoglycosides, since they inhibit protein synthesis which occurs throughout the cell cycle
What does it mean when an antibiotic is cell cycle dependent?
The antibiotic is only effective at a certain stage of the cell cycle, so that means killing of the cell is dependent on how long the drug is around (time dependent), but not on how much of the drug is around (concentration independent), since that won’t affect how many cells are at a certain stage
What adverse effects are associated with Metronidazole?
Mutagenic; Alcohol intolerance (council patient not to drink)
What is the association between β-lactams and the CSF?
They cannot cross the BBB, and they are actively pumped out; however, with CSF inflammation, β-lactams can cross due to increased capillary permeability
How do blood levels differ between aqueous, procaine, and benzathine Pen G, when given IM?
Aqueous will be rapidly absorbed and peaks high, early; Benzathine does not peak high, but has a very sustained level of release over a long time period; Procaine is an intermediate form
What adverse effects are associated with Sulfonamides?
Skin rash (can become Stevens-Johnson syndrome); Acute hemolytic anemia (with G6PDH deficiency); Kernicterus (bilirubin deposition in basal ganglia, affects newborns, not recommended in 3rd trimerster)
What adverse effect is associated with Chloramphenicol?
Bone marrow dysfunction: anemia, low incidence of pancytopenia (potentially fatal)
What adverse effects are seen with Macrolides?
GI disturbances (irritation and blockade of small bile ducts); Prolonged QT (like Quinolones) – cannot put a patient on Quinolone/Macrolide if they are taking a CCB
What adverse effects are associated with Quinolones?
CNS effects (HA, insomnia, agitation, seizures – i.e. not indicated in a patient with epilepsy);
Connective tissue disorders (should be avoided in children, tendonitis/tendon rupture);
Prolonged QT; Photosensitivity; Dysglycemia (hyper > hypo)
\What adverse effect is seen with Minocycline (Tetracycline)?
Vertigo (reversible)
What adverse effect are associated with Clindamycin?
Diarrhea, C. difficile colitis
What drug is known to compete with the efflux pump responsible for pumping Penicillin out of the CSF as well as into the urine?
Probenecid (not an abx)
What is the main indication for Vancomycin?
MRSA/MRSE – since they are resistant to β-lactams
What combination of drugs should be avoided with Cephalosporins?
Potential nephrotoxins: Aminoglycosides, Vancomycin
What administration of Penicillins is contraindicated?
Intrathecal administration since it can cause seizures
What problem can arise when giving a patient on Spironolactone a Penicillin salt preparation?
Hyperkalemia, since Spironolactone in K sparing and the Penicillin salt contains K (!)
Which antibiotics show synergism with the Aminoglycosides?
Aminopenicillins and Anti-pseudomonal Penicillins (micro lecture also included 2nd/3rd gen. Cephalosporins in this list) – there is in virto in activation of Aminoglycosides by Penicillins, so they should not be given through the same
How do amoxicillin and ampicillin differ?
Amoxicillin has better oral absorption and less diarrhea
How does excretion differ between Penicillins and penicillinase-resistant Penicillins?
Penicillins undergo active renal excretion at the PCT, so renal status should be taken into consideration; penicillinase-resistant Penicillins do not undergo urinary excretion (they have a much longer half-life)
What is the key difference between Penicillin G and V?
Penicillin V is a semisynthetic antibiotic which has an added portion to its structure that gives it acid stability (increases bioavailability)
What does the common nucleus of Cephalosporins consist of?
7-aminocephalsporanic acid: contains a β-lactam ring and a 6-membered dihydrothiazine ring
What are the extended-spectrum pens?
Antipseud pens: ticarcillin, piperacillin
What are the broad-spectrum pens?
Amino-penicillins: ampicillin, amoxicillin; their hydrophobic amino group facilitates their penetration through the outer membrane of Gram- bacteria (e.g. E. coli)
How Methacillin antibiotics inhibit β-lactamase?
Their R-group contains two methoxy groups that sterically inhibit the action of β-lactamase (these drugs are also called antistaph pens)
What are the different Penicillins and what distinguishes between them?
Penicillin G: found in different formulations (aqueous, procaine, benzathine); Penicillin V: oral administration; Methicillin: effective in Class A β-lactamase producers, includes nafcillin and oxacillin
What does the common nucleus of Penicillins consist of?
6-aminopenicillanic acid: contains a β-lactam ring and a 5-membered thiazolidine ring
What are the two main classes that antibiotics are divided up in, and what are the subdivisions associated with them?
Small molecular: chemotherapeutic agents (synthetic chemical origin, sulfonamide), antibiotic agents (microbial origin, penicillin G), semisynthetic antibiotics (chemically modified antibiotic, methicillin); Large molecular: peptide antibiotics (microbial origin, not commercialized, bacteriocins)
What is the difference between narrow-spectrum and broad-spectrum antimicrobials?
Narrow: primarily effective against one class of microbes (e.g. Gram+, penicillin G);
Broad: effective against multiple classes of microbes (e.g. Gram+/Gram-, amoxicillin)
What is the difference between bacteriostatic and bactericidal antimicrobials?
Bacteriostatic: inhibits bacterial multiplication (e.g. sulfanilamide); Bactericidal: kills bacteria (e.g. penicillin G and other β-lactam antibiotics)
What is the difference between 1st, 2nd, 3rd generation Cephalosporins?
1st gen. (e.g. cephazolin): used for Gram+ bacteria that have PBP-3 (penicillin-binding protein-3);
2nd gen. (e.g. cefoxitin, cephamycin): used in OB/GYN for anaerobe coverage, better than 1st gen. against Gram-, but less effective against Gram+, some cross BBB;
3rd gen. (e.g. ceftriaxone): even better for Gram- but worse for Gram+, crosses BBB, used in combination or alone, restricted for use on bacteria resistant to 1st/2nd gen. drugs
Which Penicillin binding proteins (PBPs) are found in microbes and which antibiotics bind to them?
PBP1: binds Penicillins; PBP2a: doesn’t bind any commercial β-lactam antibiotic (MRSAs); PBP3: binds Cephalosporins
What is meant by the terms R, MDR, and XDR in regards to drug resistance?
R: resistant to a single antibiotic, or a single class; MDR: resistant to drugs from two or more classes; XDR (extended drug resistance): MDR and R to fluroquin or amikacin (assoc. with TB)
Which Cephalosporin is considered 4th generation by some?
Cefepime: effective against Gram- and Gram+
What characterizes the pharmacodynamics of Streptomycin?
It is bactericidal for aerobic Gram- bacteria; OM active transport is required; Bacterial resistance is mediated by enzymatic mechanisms; Toxicities include ototoxicity and nephrotoxicity (!)
What uses are associated with Doxycycline?
Contraindicated in pregnancy (hepatotoxic) and young children (discolors developing teeth)
What pharmacodynamics are associated with Tetracyclines (e.g. Doxycycline)?
They are bacteriostatic, but dose-dependent (i.e. high-dose is bactericidal) and organism-dependent; They are broad-spectrum; Do not take oral formulations with milk (!), since bivalent cations (Ca2+, Mg2+) will chelate the Doxycycline and prevent its absorption
What does Tetracycline activity and resistance center around?
Membrane pumps that either pull Tetracycline into the cell or pump it out
How does the mechanism of action differ between that of Tetracyclines and Aminoglycosides?
Tetracyclines also inhibit proteins synthesis at the 30S ribosome subunit, but they do so by preventing tRNA to mRNA binding (vs. preventing tRNA to 30S binding for Aminoglycosides)
How does Gentamycin differ from Streptomycin?
Gentamycin is much more broad-spectrum
What are the uses of the Aminoglycosides?
In combination with β-lactams: Gentamycin has a 4x synergy with 4th gen. Penicillins or 2nd/3rd gen. Cephalosporins (synergy occurs because Gentamycin causes membrane damage which allows the pens/cephs to enter more easily)
What is the therapeutic use of Methacillin?
Sepsis, endocarditis, and meningitis caused by susceptible S. aureus strains
What is the mechanism of action of the Aminoglycosides?
Streptomycin: inhibits protein synthesis by preventing binding of tRNA to the bacterial 30S ribosome subunit
What is the oral formulation of Methacillin?
Dicloxacillin
What uses are associated with Macrolides?
Substitute for Penicillins in pen allergic patients; covers major bacterial causes of walking pneumonia (e.g. Mycoplasma pneumoniae)
What are the most overprescribed antibiotics?
Cephalosporins (followed by Fluoroquinolones)
Which Fluoroquinolone does affect S. pneumoniae?
Moxifloxacin
What are the uses of Ciprofloxacin?
UTI, STD, RTI (not useful when caused by S. pneumoniae, but good for hospital acquired RTIs)
What pharmacodynamics are associated with Ciprofloxacin?
Dose-dependent bactericidal for Gram+/-; excellent bioavailability except in CSF
What is the mechanism of action of Fluoroquinolones (e.g. Ciprofloxacin)?
Interferes with topoisomerase II and IV
What are examples of extended spectrum Macrolides?
Clarithromycin, Azithromycin
How does Tigecycline differ from other Tetracyclines?
It is refractory to the influx/efflux pumps required for Tetracycline activity and resistance
What pharmacodynamics are associated with Macrolides?
Bacteriostatic (but bactericidal in high doses) for selected Gram+/Gram-;
What is the mechanism of action of the Macrolides (e.g. Erythromycin)?
Binds to the 50S subunit and causes early peptide chain release
What new drug is part of the Glycopeptide classification?
Dalbavancin
What elicits the strength of an antibiotic?
Its killing curve: the steeper the slope, the more effective it is
In Dilution AST, what is meant by the MIC and MBC?
MIC: first test tube, in a serial-dosage test, that is not cloudy with bacteria (dose is bacteriostatic);
MBC: first test tube that shows almost no growth when plated on a Petri dish (dose is bactericidal) – MBC dosage is important in immunocompromised patients who can’t finish off the remaining bacteria left by the MIC dose (also in patients with endocarditis and osteomyelitis)
What is the purpose of Dilution AST?
Antibiotic susceptibility test – determines which antibiotics will be effective against a certain bug isolated from a patient and grown in culture
What uses are associated with Sulfamethoxazole?
In combination with Trimethoprim, which is a non-competitive inhibitor dihydrofolate reductase (same metabolic pathway) – both can be used at low doses to minimize adverse effects; it is formulated as co-trimoxazole (TMP/SMX, Bactrim, Septra) and used for UTI
What pharmacodynamics are associated with Sulfamethoxazole?
Bacteriostatic; Broad-spectrum; Adverse reactions are common (fever/rash/photosensitivity)
What is the mechanism of action of Sulfonamides (e.g. Sulfamethoxazole)?
Competitive inhibitor of PABA in folate synthesis (tetrahydrofolate is required for deoxynucleotide synthesis, bacteria cannot absorb folate but must synthesize it) – perhaps the only antibiotic that functions by inhibiting intermediate metabolism of bacteria
What is the mechanism of action of the Glycopeptides (e.g. Vancomycin)?
Blocks cell wall transpeptidization by binding to the D-ala/D-ala terminus of the peptidoglycan pentapeptide thus preventing the incorporation of NAG/NAM subunits into the peptidoglycan matrix (compare to the β-lactams which inhibit the enzyme DD-transpeptidase, and thus prevent peptidoglycan cross-linking)
What pharmacodynamics are associated with Vancomycin?
Bactericidal (like β-lactams); Narrow-spectrum: Gram+; rapid infusion toxicity: hypotension; must monitor oto/renal functions
What uses are associated with Vancomycin?
MRSA caused sepsis but also used orally for C. dif intestinal toxicosis
If an object contains 1x106 bacteria and an disinfectant is applied that kills 90% of the bacteria every 10 min., how many will be left after 40 minutes?
Every 10 minutes, 10% of the original number will be left, so after the first 10 min, 1x105 bacteria are left; the exponent decreases by 1 every 10 minutes, so after 40 min. there will be 1x102 left (i.e. 100)
What is an example of “cidal-static” antagonism?
Giving Penicillin and Tetracycline together: the Tetracycline will prevent growth which will make the effects of Penicillin (i.e. killing bacteria that are synthesizing new cell walls) useless
What are examples of Clavulenic acid drugs?
Clavulanate, Sulbactam, Tazobactam
What is an example of a suicide molecule and what is its mechanism of action?
Clavulenic acid: has a high affinity for β-lactamases produced by various microbes, which they irreversible bind and inactivate, thus allowing other β-lactam antibiotics to do their job undisturbed – these are not effective on MRSA since their resistance is based on PBPs, not β-lactamase
How are the S and R breakpoints used?
By looking at the killing curve for a particular microbe/antibiotic combination and finding the concentration of antibiotic that is necessary to kill 80% of the microbe; if this concentration falls before the predetermined S breakpoint, the microbe will be susceptible; if it falls between the S and R breakpoints, it will be intermediate; and if it falls after the R breakpoint, it will be resistant
What is the use of breakpoints regarding Dilution AST?
It determines the SIR: is a bacterial isolate Susceptible, Intermediate, or Resistant to a particular antibiotic
Why are Bactrim/Septra used prophylacticly in females with recurrent UTIs?
The normal vaginal secretions are acidic and will cause ion trapping of the drugs; this will ensure that an infection in the making will be killed before changing the vaginal pH
How do Fluoroquinolones cause possible CNS effects?
Through their effects on GABA inhibition (also true for NSAIDs)
Which substances can cause a decreased absorption of Fluoroquinolones?
Antacids containing vitamins (Zn, Mn, Mg, Fe) and milk; this applies to Tetracyclines as well; administer the drugs 2-4 hours apart
Because of their mechanism of action of inhibiting DNA gyrase (Topo II) and Topo IV, are Fluoroquinolones cell cycle specific?
NO! – effective in static cultures
What is the current drug of choice for chronic bacterial prostatitis?
Fluoroquinolones
What are the two most common Fluoroquinolones?
Ciprofloxacin (Cipro), Levofloxacin (Levaquin)
What should patients be counseled to do when taking Bactrim/Septra and why?
Maintain proper hydration since irreversible renal impairment can occur due to crystalluria (really only a problem in patients with renal disease)
Which enzymes are inhibited by Sulfonamides and Trimethoprim, respectively?
Sulfonamides: Dihydropteroate synthetase (PABA --> folic acid), enzyme not present in humans;
Trimethoprim: Dihydrofolate reductase (folic acid --> tetrahydrofolic acid), enzyme present in humans, but antibiotic has a much higher affinity for the bacterial enzyme than the human form
What is the absorption/excretion associated with Sulfonamides/Trimethoprim (Bactrim/Septra)?
Oral absorption, renal excretion (good for UTI)
Why is Trimethoprim not effective in bacterial prostatitis?
Although it was thought to be effective due to ion trapping in the prostate fluid, the pH of this fluid in bacterial prostatitis is much higher, diminishing the ion trapping effect and reducing effectiveness of the drug
What is the danger of giving theophylline with Fluoroquinolones?
Theophylline levels can be increased due to decreased hepatic catabolism (can lead to a decrease in cAMP degradation which can have cardiac effects, such as prolonged QT); Theophylline may also potentiate the GABA inhibition effects already caused by the Fluoroquinolones
What are some of the indications for Tetracyclines?
Rickettsial infections; Mycoplasma pneumoniae; Chlamydiae; Acne
What is special about the elimination of Doxycycline (part of Tetracyclines class)?
It does not undergo renal clearance
What is the mechanism associated with increased insulin release seen in patients taking Fluoroquinolones?
Fluoroquinolones bind to a sulfonylurea receptor and cause closure of K channels --> this leads to depolarization of the cell, which will cause Ca channels to open --> the influx of Ca is the stimulus for insulin release (under normal circumstances this process is initiated by glucose influx into the cell, raising the ATP/ADP ratio)
What characterizes the glucose dysregulation seen with Fluoroquinolones?
Initial hypoglycemia due to increased insulin release; followed by hyperglycemia due to some unknown mechanism; the hyperglycemia can be exacerbated by glucocorticoid steroids –
people most prone to dysglycemia are diabetics with renal impairment receiving insulin/oral hypoglycemics
Why are the Aminoglycosides ototoxic and what other drugs are known to exacerbate these effects?
The half-life of the drug is much longer in otic fluid – ototoxicity is exacerbated by loop diuretics! (Furosemide) and Vancomycin
In what class of bacteria is Metronidazole effective?
Anaerobes
What is a key difference between Clindamycin and Chloramphenicol?
Clindamycin does not get into the CNS
What potentially fatal side effect is associated with the hepatic elimination of Chloramphenicol?
Gray baby syndrome: pre-mature babies/neonates have undeveloped hepatic conjugation and renal elimination which can lead to toxicity; these toxicities include vomiting, decreased respiration, green color of loose stools, hypothermia, cyanosis, and possibly death
What is special about the distribution of Chloramphenicol?
It has a uniquely high CSF distribution
Which antibiotics have a mechanism of action that involves interfering with the bacterial 50S ribosomal subunit?
Macrolides (Erythromycin, Azithromycin, Clarithromycin), Clindamycin: steric inhibition;
Chloramphenicol: direct inhibition
What factors can exacerbate the nephrotoxicity seen with the Aminoglycosides?
Hydration status, Indomethacin, Vancomycin, some β-lactams
What drug is used for the treatment of Legionnaires disease?
Azithromycin (Macrolide abx)
Although Macrolides are associated with GI disturbances, when a patient ceases to take the drugs and the GI effects continue, what underlying problem should be taken into consideration?
Pseudomembranous colitis due to C. difficile
Which class of antibiotics is rapidly bactericidal and has a post-antibiotic inhibitory effect?
Aminoglycosides – can be given once daily since it is cell cycle independent and because of its post-antibiotic effect (peak-trough regimen)
What is the mechanism of action of Metronidazole?
Pro-drug; active metabolite reacts with functional groups of DNA and protein causing DNA strand breaks (causes mutagenicity, thus contraindicated in 1st trimester)
What adverse effects are associated with Metronidazole?
HA, urine discoloration, mutagenicity, alcohol – “Antabuse” reaction
How can antibiotics be responsible for lowered plasma contraceptive levels?
By killing some bacteria in the intestine, bacterial deconjugation of eliminated contraceptives is reduced; this means that less contraceptive is reabsorbed after bile elimination
By what process can plasmids be transformed into bacteria that are not typically transformable?
Through electroporation (add plasmid to bacteria and pass a current at high voltage through them)
Where do antibiotic resistance genes come from?
Often from soil microorganisms (they have them to protect themselves)
What are the two general types of phages?
Lytic (virulent): always kill the host cell; Lysogenic (temperate): may go through lytic cycle, but can sometimes form a lysogen (dormant prophage becomes part of bacterial chromosome) – if a phage that contains a gene encoding for a toxin becomes a lysogen, it can confer the toxic gene to the bacteria, thus increasing its virulence
Genes associated with what function are often carried in bacteriophages?
Genes that encode for toxins (transfer process is called specialized transduction)
What is the case in an Hfr strain of bacteria?
Hfr stands for high frequence of recombination – in an Hfr strain the F plasmid has integrated itself into the bacterial chromosome, meaning that the entire chromosome is transferred during conjugation
How does bacterial conjugation occur?
The donor cell and recipient join via a pilus (the donor cell must contain the F plasmid in order to form the pilus); the plasmid in the donor cell is then nicked and transferred in its single-stranded form; once it enters the recipient, strand replication will occur and the plasmid is reformed
By what mechanisms does multiple drug resistance occur?
De novo mutations; Plasmid acquisition
What are the three different mechanisms for bacterial gene transfer?
Transformation: transfer of “naked” DNA (no protein coat); Transduction: DNA carried by a bacterial virus (phage); Conjugation: requires direct cell-cell contact where a plasmid is sent through a pilus
Which bacterial gene transfer mechanism plays a major role in drug resistance transfer?
Conjugation
What characterizes transformation?
Bacteria lyse in stationary phase releasing their contents; DNA from lysed bacteria is bound by specific surface proteins and is transported into other cells through the membrane; once inside, if the DNA is a chromosomal gene it will undergo homologous recombination only if the cell it entered is of the same species (if it is a different species, this cannot occur); however, if the DNA is a plasmid, it can cross species barriers – this mechanism can be used to transfer useful traits
What does generalized transduction refer to?
When a phage goes through the lytic cycle, instead of packaging its own DNA, it packages a part of the host DNA
What are the locations on Salmonella’s genome called where the Type III secretion apparatuses are found?
Pathogenicity islands (large AT heavy regions)
How does Salmonella typhimurium get to its target organs?
It adsorbs to and travels through the gut epithelium (via a Type III secretion apparatus); once inside Peyer’s patches it uses macrophages to travel in the body (again via the Type III secretion apparatus); eventually it grows in the spleen and liver
By what mechanism does Salmonella switch between the production of different flagellins?
Normally, a promoter sequence allows for transcription of H2 flagellin along with a repressor for H1 flagellin --> the promoter sequences is flanked by transposable elements and can become inverted --> when this occurs, the promoter becomes inactive and transcription of H2 and the H1 repressor is halted --> this in effect allows for the transcription of H1 and the switch is complete
What is meant by antigenic variation?
Use of alternative structural proteins to form identical structures (this allows the micro be form a protein that is not cross-reactive, i.e. the immune system doesn’t respond to it yet) – e.g. flagellin protein in Salmonella
What are transposable elements?
DNA segments that can move to other sites – a transposon is a type of complex transposable element (these can be capable of moving antibiotic resistance genes)
What are the modified smooth muscle cells that surround capillaries?
Pericytes
What two factors are important in maintaining blood pressure?
RAAS and [Na]
What is a common cause of malignant hypertension and what sign is associated with this disease?
Cause: renal failure; Sign: Retinal hemorrhages and exudates with or without papilledema
What causes hypertension in patients?
95% is idiopathic (essential hypertension); 5% is malignant (death in ~2 years)
What are the three patterns of arteriosclerosis and what describes each?
Arteriolosclerosis: hardening of the arteries by hyaline deposits or blood vessel hyperplasia ; Monckeberg medial calcific sclerosis: calcification of the internal elastic lamina;
Atherosclerosis: thickening of the intima with fatty streaks and plaques
Where do 90% of berry aneurisms occur?
In the anterior circulation --> cause subarachnoid hemorrhage
Which blood vessels are most easily penetrated by tumors and inflammatory processes?
Veins
What complication is problematic with arteriovenous malformations (AVM)?
The pressure of the arterial system is transmitted directly to the venous system, causing hemorrhage
What are the layers of a blood vessel from the inside outwards?
Tunica intima (single layer of endothelial cells) --> Internal elastic lamina --> Tunica media (smooth muscle) --> External elastic lamina --> Tunica adventitia (connective tissue with nerve fibers and vasa vasorum)
What are the principle points of physiologic resistance to blood flow?
Arterioles
What type of arteriolosclerosis is characteristic of malignant hypertension?
Hyperplastic arteriolosclerosis: “onion-skinning” of the blood vessel
What clinical signs are associated with a thoracic aneurism?
Respiratory difficulties; Difficulty swallowing; Persistent cough; Pain caused by erosion of bone; Cardiac disease
What characterized syphilitic (luetic) aneurisms?
Tertiary stage shows predilection for small vessels; Inflammation begins in the aortic adventitia, particularly the vasa vasorum; Ischemic injury of the tunica media
Where do abdominal aneurysms usually occur?
Below the renal arteries but above the bifurcation of the aorta; a resulting thrombus can clog nearby arteries (renal, mesenteric, etc.)
What terms describe the morphology of different aneurysms?
Saccular (spherical): involve only a portion of the wall, partially/completely filled with thrombus;
Fusiform: involve a long segment of the vessel (e.g. large segment of the aorta)
What is a mycotic aneurism?
An aneurism caused by infection of a major artery that weakens the vessel wall
What is the difference between true and false aneurysms?
True: bound by arterial wall components (atherosclerotic, syphilitic, congenital, left ventricular);
False: breach of vascular wall (hematoma, post-MI rupture, leak at anastomosis of vascular graft)
Which humoral factors are involved in blood vessel constriction and dilation, respectively?
Constriction: Angiotensin II, Catecholamines, Thromboxane, Leukotrienes, Endothelium;
Dilators: Prostaglandins, Kinins, NO
What neural factors are associated with blood vessel constriction and dilation, respectively?
Constriction: α-adrenergic; Dilation: β-adrenergic
What is the major morphologic characteristic of benign nephrosclerosis?
Hyaline arteriolosclerosis: seen in elderly patients, hypertensive patients, and diabetics
What are the main categories of causes associated with vasculitis?
Direct invasion of vascular walls by infectious pathogens; Immune-mediated mechanisms; Physical and chemical injury (irradiation, mechanical trauma, toxins)
Which disease is associated with weakening of pulses in the upper extremities (“pulseless disease”)?
Takayasu Arteritis: females<40, granulomatous vasculitis, mainly affects aorta and pulmonary arteries, adventitial mononuclear infiltrate to intense mononuclear inflammation in the media, coldness/numbness of fingers, ocular disturbances – involves HLA-A24/B25/DR2
What signs and symptoms are associated with Giant Cell Arteritis?
Fever, fatigue, weight loss; Facial pain or HA along the course of the superficial temporal artery; Diplopia to transient/complete vision loss – diagnosed by vessel biopsy and histologic confirmation
Which arteries are principally involved in Giant Cell Arteritis?
Temporal, vertebral, ophthalmic arteries (medical emergency); can also be associated with a thoracic aortic aneurysm
Which diseases are associated with AECA?
Lupus; Kawasaki disease
Which diseases are associated with particular forms of ANCA?
c-ANCA: Wegener’s granulomatosis; p-ANCA: Microscopic polyangiitis, Churg-Strauss
What antibodies are commonly found in the serum of a patient with vasculitis?
Anti-neutrophil cytoplasmic antibodies (ANCA); Anti-endothelial cell antibodies (AECA)
What distinguishes Type A and Type B aortic dissections?
Type A: involves ascending aorta (and possible descending as well); Type B: involves only the descending aorta (distal to subclavian artery)
What clinical presentation is associated with vasculitis?
Fever, myalgias, arthralgias, malaise
What are some complications associated with an aortic dissection?
Cardiac tamponade, aortic insufficiency, MI, extension of dissection into great arteries of neck, rupture (=most common cause of death) – 65-75% survival
What clinical signs are associated with Kawasaki Disease?
Erythema of palms and soles, skin rash with desquamation and enlargement of cervical lymph nodes; 20% may develop cardiac sequelae – necrosis and inflammation affects entire thickness of vessel wall; ASA and IV γ-globulin have reduced fatalities to 1%
What clinical picture is seen with Buerger’s disease?
Adult smoker; severe pain with skin lesions
What is Buerger’s disease also known as and what characterizes it?
Thromboangiitis obliterans: segmental, thrombosing, acute and chronic inflammation of the medium/small arteries (usually tibial/radial arteries, but can extend to associated nerves/veins)
What causes Wegener’s granulomatosis and what clinical signs are associated with it?
c-ANCA; Seen mostly in adults; Associated with upper respiratory lesions and renal lesions
What pathology is associated with Wegener’s granulomatosis?
Acute necrotizing granulomas of the upper/lower respiratory tracts; Necrotizing/granulomatous vasculitis of small/medium-sized vessels; Focal necrotizing/crescentic glomerulitis
What clinical signs are associated with Microscopic Polyangiitis?
Hemoptysis, abdominal pain, muscle weakness/pain, hematuria/proteinuria, palpable purpura, leukocytoclasia (infiltration of the media with neutrophils) – skin biopsy is diagnostic, p-ANCA will be positive in 70% of cases, caused by immunologic reaction (remove offending agent)
What is the key difference between PAN and Microscopic Polyangiitis?
In PAN the medium-size arteries are involved, whereas with Microscopic Polyangiitis the smaller vessels (arterioles, capillaries, venules) are involved – both are segmental transmural necrotizing inflammations
Which systemic vasculitis presents as a segmental transmural necrotizing inflammation?
Polyarteritis Nodosa (PAN): fibrous thickening of vessel wall, lumen may be thrombosed, all stages of activity may exist in different vessels or in the same vessel
What is the clinical picture of PAN?
Young adult, remittent/episodic with long symptom-free intervals, Hepatitis B association (30%), hypertension, melena (due to vascular lesions in GI tract), peripheral neuritis – no association with ANCA, Tx with corticosteroids and cyclophosphamide
What characterizes Kawasaki Disease?
Mucocutaneous lymph node syndrome in young children (may result from immune reaction); Kawasaki may lead to arteritis of the coronary arteries (which can lead to MI)
What is thrombophlebitis and what causes it?
Venous inflammation with possible thrombus formation (can lead to pulmonary embolism) most commonly in the deep leg veins; can be caused by cardiac failure, neoplasia, pregnancy, obesity, prolonged bed rest, and genetic hypercoagulability syndromes
How do cavernous hemangiomas differ from capillary hemangiomas?
They are less common; Less well circumscribed; Involve deeper structures; May not regress – there is an association with von Hippel-Lindau disease (genetic condition involving tumors in vascular rich parts of the body)
What is a capillary hemangioma?
Largest single type of vascular tumor; found in infants, usually fade with age
What is angiomatosis?
Multiple angiomas involving large segments of various organs
What signs/symptoms are associated with lymphangitis?
Dilated lymphatics filled with exudate (due to group A β-hemolytic strep); Painful subcutaneous red streaks extending along the course of lymphatics with painful enlargement of regional lymph nodes
What causes SVC and IVC syndromes and what signs are associated with them?
SVC syndrome: neoplasia compressing SVC, associated with dusky cyanosis, dilation of veins;
IVC syndrome: neoplasia or thrombi, associated with edema of the legs, distension of superficial collateral veins of the lower abdomen and massive proteinuria
What two clinical tests can be done to check for thrombophlebitis?
Trousseau sign; Homan sign: passive dorsiflexion of the foot elicits sharp calf pain
What is the Raynaud phenomenon?
Ischemic attacks of the extremities: cold-induced vasoconstriction of digital arteries, precapillary arterioles, and cutaneous arteriovenous shunts (color changes: white --> blue --> red)
What clinical picture is associated with Raynaud phenomenon?
Median age: 14 years old; may have family history; atrophy of skin, subcutaneous tissues, and muscles – must exclude secondary Raynaud phenomenon in case it is due to other diseases (e.g. SLE, atherosclerosis, Buerger’s)
What causes varicose veins?
Prolonged, increased intraluminal pressure (e.g. due to long periods of standing, obesity, pregnancy) causes loss of vessel wall support, especially in the superficial veins of the upper/lower legs of people over 50 years of age
What benign, painful tumor is often found under the finger nails?
Glomus tumor (glomangioma): arises from modified smooth muscle cells of the glomus body
What is angiosarcoma?
Malignant endothelial neoplasm found mostly in older adults; can be due to arsenic, Thorotrast, PVC, and radiation exposure – poor outcome
Which blood vessel abnormality is an intermediate between hemangioma and angiosarcoma?
Hemangioendothelioma
What virus are 95% of Kaposi’s sacromas associated with?
Herpesvirus 8 (HHV-8)
What are the four types of Kaposi’s sarcoma?
Classic/European: not associated with HIV; African/endemic: lymphadenopathy, aggressive; Transplant/immunosuppressed-associated: aggressive; AIDS-associated: most common
What is Bacillary Angiomatosis?
Vascular proliferation due to opportunistic infections (Bartonella family) in immunocompromised people – also known as Cat Scratch Fever and Trench Fever
What are the vascular ectasias and what diseases are they associated with?
Nevus flammeus (birthmark/port-wine stain): Sturge-Weber syndrome; Spider telangiectasia: pregnancy and cirrhosis; Hereditary hemorrhagic telangiectasia: Osler-Weber-Rendu disease
What is a pyogenic granuloma?
Lobular capillary hemangioma: polypoid rapidly growing exophytic red nodule attached by a stalk (can develop after trauma)
What are capillary lymphangiomas?
Network of endothelium-lined lymph spaces (absence of blood cells); seen with Turner syndrome
What characterizes LDL?
Originate from membrane surface of cells; Composed of triglycerides (9%) and cholesterol (60%); Normally removed from the blood by receptor-mediated uptake into the liver and other tissues; Closely associated with coronary artery disease (CAD)
What side effects are associated with statins?
Muscle damage (rhabdomyolysis and myalgias); Acute renal failure; Increased serum liver transaminases (liver failure is rare); Bloating, diarrhea, constipation
How does limiting cholesterol biosynthesis by HMG CoA reductase reduce LDL?
A reduction in cholesterol biosynthesis leads to a decrease of intracellular cholesterol levels available for bile acid synthesis; As a compensatory mechanism, the liver upregulates its LDL receptors, thus removing LDL from the blood – IDL and VLDL remnants are also removed
How do HMG CoA reductase inhibitors function?
The inhibits the rate-limiting step in cholesterol biosynthesis; also, they modify endothelial function, inflammatory responses, plaque stabilization, thrombus formation, and reduce plaque formation
What is the “gold standard” drug class for lowering plasma cholesterol?
HMG CoA reductase inhibitors (i.e. –statins)
Which has a greater impact on cardiovascular events, increased LDL or reduced HDL?
Reduced HDL
What characterizes HLD?
Produced in liver; Composed of triglycerides (8%) and cholesterol (17%); Transport cholesterol from periphery to liver; Inversely correlated with CAD
What are the risk factors for coronary artery disease for LDL, HDL, and total cholesterol levels?
LDL<130; HDL>40; Total cholesterol<200
What characterizes chylomicrons?
Originate in the small intestine; Composed of dietary triglycerides (90%) and cholesterol (3%); Chylomicron triglycerides are broken down by lipoprotein lipase leaving a cholesterol-rich chylomicron remnant which is taken up by the liver
What characterizes VLDL?
Originate in the liver; Composed of triglycerides (55%) and cholesterol (dietary/de novo – 16%); Metabolism of triglycerides by lipoprotein lipase converts VLDL to LDL
What is the mechanism of action of Cholestyramine and Colestipol?
They are ion exchange resins that bind to bile acids in the small intestines preventing their reabsorption --> increased conversion of liver cholesterol to bile acids --> de novo synthesis can’t keep up --> upregulation of liver LDL-receptors --> decrease in plasma LDL
What is the function of fibric acid derivatives (e.g. Clofibrate)?
Lower triglycerides by stimulating lipoprotein lipase activity to break down plasma triglycerides from lipoproteins (especially from chylomicrons and VLDL)
Which drug appears to reduce plaque volume?
Synthetic HDL
What kind of drug is Anaretrapib?
It is an investigational drug that functions as a CEPT inhibitor (CEPT mediates the transfer of cholesterol esters and triglycerides between lipoproteins); it is supposed to markedly increase HDL and decrease LDL
What is the mechanism of action of Ezetimibe?
It inhibits the uptake of cholesterol in the intestinal lumen by acting at the brush border to reduce cholesterol transport across the cell wall – can be used in combination with statins (e.g. Vytorin)
How does Colesevelam differ from other bile acid binding drugs?
It is a hydrogel polymer, causing it to have less incidence of GI disturbances and it does not appear to reduce absorption of acidic drugs as do the resins
What side effects are associated with the bile acid binding products?
Significant GI disturbances (constipation/steatorrhea); Bind certain acidic and hydrophobic drugs in the GI tract inhibiting their absorption (e.g. digitoxin, phenobarbital, tetracyclines); They can also inhibit intestinal absorption of fat-soluble vitamins
What contraindications are associated with statins?
Pregnancy and lactation; Liver disease and cholestasis
What drug interactions are associated with statins?
Some statins are metabolized by CYP3A4, so inhibitors of 3A4 should be avoided (e.g. cyclosporine, amiodarone, ketoconazole, erythromycin, HIV protease inhibitors); also concurrent use of statins with Gemfibrozil can increase the risk of myopathy
Which CYP enzymes are responsible for the metabolism of common statins?
3A4: Lovastatin, Simvastatin; Partial 3A4: Atorvastatin, Fluvastatin; Partial 2C9/2C19: Rosuvastatin, Fluvastatin; NOT metabolized by CYP enzymes: Pravastatin
What side effects are associated with Clofibrate?
Increased risk of gallstones, increased risk of mortality – not prescribed much anymore
Which combination therapy has the most potential for lowering LDL/triglycerides while raising HDL?
Statin/Niacin
What side effects are associated with Niacin?
Intense flushing and itching; Hepatotoxicity; GI disturbances – an extended release formula (Niaspan) has fewer side effects
What is the mechanism of action of Niacin (Vitamin B3)?
It decreases the breakdown of triglycerides in adipose tissue by inhibiting lipolysis; this decreases the supply of FFAs for the synthesis of VLDL which will in turn decrease the formation of LDL; it also increase HDL
What is Omacor used for?
Reduce triglycerides and increase survival post-MI; It is an Omega-3 polyunsaturated fatty acid preparation
What is the mechanism of action of Fenofibrate?
Similar to Gemfibrozil but functions by inhibiting AcCoA carboxylase; this decreases fatty acid synthesis and increases LDL uptake by the liver – used to reduce CAD in patients with DM-II
What is the mechanism of action of Gemfibrozil?
Stimulates lipoprotein lipase on the endothelium (lowering triglycerides); Inhibits the synthesis of apolipoprotein B (a carrier molecule for VLDL, IDL, and LDL) thus decreasing plasma LDL;
It also increases HDL modestly
What are the main major arteries in which atherosclerosis is a clinical concern?
Aorta, carotid a., iliac a. (for medium-sized arteries: coronary a., popliteal a.)
Where in the plaque do lymphocytes accumulate?
In the shoulders of the plaque
What happens with small-artery atheromas?
Ischemic injury
What is a key characteristic of atherosclerosis in regard to the vessel wall?
Intimal lesions and weakening of the underlying media
What are some less common consequences of atherosclerosis?
Sudden cardiac death, Mesenteric occlusion, Chronic ischemic heart disease, Ischemic encephalopathy, Large artery atheromas, Aneurysms
What are the most dangerous plaques?
Thrombus plaques
How does a normal artery progress to a plaque?
Either directly or via a fatty streak
What is the difference between stenosis and occlusion?
Occlusion is by definition a 100% closure of the vessel, whereas stenosis is by definition less than 100% closed off
What is the difference between an eccentric and a concentric plaque?
Eccentric plaques take up one side of the vessel, whereas concentric plaques go around the entire vessel
What is a major concern with fibrous plaques in the aorta?
Thinning of the tunica media leading to aneurysm
What accumulates into class III lesions?
Oxidized LDL and cholesterol – potentiated by hypercholesterolemia
Which segment of aorta is far more susceptible to aneurysmal dilatation due to atherosclerosis?
Abdominal aorta
What are the key characteristics of atherosclerosis in regard to plaque formation?
Intimal thickening and lipid accumulation
What do derivatives of cigarette smoking in circulation lead to?
Accelerated plaque formation
What causes fatty steaks to form around ostia (entrance from one vessel into the next)?
Turbulence – these fatty streaks are called adaptive thickenings (they do not disturb blood flow)
What is contained within fatty streaks?
T lymphocytes; Extracellular lipid (in smaller amounts than in plaques) – fatty streaks are present in all people over age 10
What is a key difference between class III and IV lesions?
Class IV lesions (atheromas) have a core of extracellular lipid whereas Class III lesions have small extracellular lipid pools
How do foamy macrophages end up inside plaques?
Circulating monocytes invade into tissues, including plaques, where they differentiate into macrophages (these macrophages take up LDL and become foamy)
What are the American Heart Association atherosclerosis classification categories?
I: initial lesion of isolated foam cells; II: fatty streaks; III: intermediate lesion (raised, causes stenosis); IV: Atheromas (introduce an extracellular matrix of lipids); V: fibroatheromas (progressive fibrous cap formation with smooth muscle); VI: complicated lesions (thrombus deposition, ulceration)
Which AHA classification categories have the ability to stop progressing?
I and II
Where do fibrous plaques cause the greatest frequency of disease?
Lower abdominal aorta > Coronary a. > Popliteal a. > Internal carotid aa. > Circle of Willlis
What are the three principle microscopic components of atherosclerotic plaques?
Cells: smooth muscle cells, macrophages, leukocytes (recruited by VCAM-1); ECM: collagen, elastic fibers, proteoglycans; Intracellular- and extracellular lipid
Which cell in a plaque is most responsible for causing pathogenesis?
Smooth muscle cell (especially the ones recruited from the media by PDGF)
How can neovascularization of a plaque be problematic?
If it receives its blood supply from the lumen of the main vessel and from the vasa vasorum, the pressure in the vessel can become a problem and cause hemorrhaging of the plaque, weakening the fibrous cap
How does LDL become oxidized?
Through its exposure to free radicals (antioxidants may help stem the development of raised lesions)
What factors are responsible for the pathogenesis of atherosclerosis?
Endothelial injury, Inflammation, Lipids, Smooth muscle cells
What are modifiable risk factors of atherosclerosis?
Hyperlipidemia, HTN, cigarette smoking, Diabetes Mellitus; also Lipoprotein A, trans-fat intake, Chlamydia pneumoniae, excessive alcohol intake
What are the constitutional risk factors of atherosclerosis?
Age (older), sex (male), genetics (e.g. homocystinuria)
What is the term used when a thrombus creates a new lumen?
Recanalization
What is the most feared complication of an atherosclerotic plaque?
Superimposed thrombosis (due to disrupted lesions leading to the exposure of highly thrombogenic substances – i.e. smooth muscle cells, etc)
What is found in the “shoulder” of atherosclerotic plaques?
Macrophages, Smooth muscle cells, T-lymphocytes
What is located deep to the fibrous cap?
Necrotic core containing a disorganized mass of lipids – if the cap thins and the core is exposed, a thrombus can form
What variables can make atherosclerosis worse?
Neovascularization at the periphery of lesions; Dystrophic calcification (increased risk of coronary events)
What layer is always the one that thickens in atherosclerosis?
Intima (NOT the media or adventitia)
What vessel wall layer is affected by Mönckeberg Medial Calcific Sclerosis?
Media – as opposed to the intima in atherosclerosis
What are the two key determinants of the early stages of atherosclerosis?
Serum cholesterol concentrations; Smoking
What should risk-factor evaluation and prevention start in regards to atherosclerosis?
Childhood
What players in atherosclerosis can produce PDGF?
Platelets attached to damaged endothelium; Smooth muscle cells recruited into plaque; Macrophages – PDGF causes smooth muscle cells to migrate into the plaque
Can atherosclerosis be initiated without endothelial damage?
Yes, for example through VCAM-1 induction
What are mimickers of vasculitis and how do they present?
Subacute bacterial endocarditits (SBE): new heart murmur, splinter hemorrhages, unusually high fever; Infection: liver dysfunction (Hep C), drug abuse (HIV, Hep C, cocaine), high risk sexual activity (HIV); Cancer: history of cancer; Cholesterol emboli: necrosis of lower extremity digits
What is the treatment for PAN?
Prednisone and Cyclophosphamide (Cytoxan)
What is associated with PAN?
Hepatitis B (PAN occurs within six months of Hep B infection) – patients are ANCA negative
What signs should be suspect of PAN?
Men in 40s/50s; Palpable purpura; Peripheral neuropathy; Glomerulonephritis: red cell casts, proteinuria – should biopsy active site for diagnosis (angiogram: beating with aneurysms)
What are the clinical features of Polyarteritis Nodosa (PAN)?
Mononeuritis multiplex: transient wrist drop/foot drop; Focal necrotizing glomerulonephritis; Palpable purpura; Arthralgias; Myalgias; Abdominal claudication; Abnormal liver tests; MI; Seizure, stroke; Jaw claudication; Testicular pain; Retinal hemorrhage – basically anywhere you find medium/small vessels
What is the only way to confirm the diagnosis of Giant Cell Arteritis and what is the treatment?
Temporal artery biopsy (big piece), should also have high ESR; Treatment: Prednisone (immediately), Aspirin (helps prevent blindness)
What symptoms are associated with Giant Cell Arteritis (Temporal Arteritis)?
Superficial temporal headache (can’t wear hat/glasses/comb hair), jaw claudication, diplopia, blindness: acute or transient (amaurosis fugax), polymyalgia rheumatica (pain/stiffness in proximal muscle groups), fever or cough (can present by themselves), weight loss, fatigue
What types of vasculitis are associated with large, medium, and small vessels, respectively?
Large: Takayasu’s Arteritis, Giant Cell Arteritis; Medium: PAN, Kawasaki’s disease, Buerger’s disease, CNS vasculitis; Small: Hypersensitivity vasculitis, Cryoglobulinemia, Henoch-Schönlein Purpura, Wegener’s Granulomatosis, Microscopic Polyangiitis, Churg-Straus Vasculitis
What is claudication?
Attacks of lameness and pain (mainly in the calf muscles) upon exertion – caused by ischemia
What signs should be suspect of vasculitis?
Multisystem disease, unexplained constitutional signs/symptoms, palpable purpura, gangrene, claudication, Raynauld’s, livedo reticularis, glomerulonephritis, mononeuritis multiplex, myalgias/arthralgia/arthritis, intestinal angina, testicular pain
What clinical manifestations are associated with Wegener’s granulomatosis regarding the lungs and kidneys?
Upper respiratory tract symptoms: sinusitis, nasal discharge, epistaxis, nasal ulcerations, nasal septum perforations, saddle-nose deformity, oral ulcers, otitis media, subglottic stenosis (stridor); Lower respiratory tract symptoms: asymptomatic, chronic cough, alveolar hemorrhage, pneumonitis, respiratory insufficiency, pleuritis;
Renal symptoms: Pauci-immune necrotizing glomerulonephritis (i.e. no immune-complex deposition), asymptomatic, active urine sediment: hematuria, pyuria, proteinuria, casts
What is the treatment associated with Wegener’s?
Generalized/Limited WG: Prednisone, Cytoxan; Milder WG: MTX or Cyclosporin –
Patients on Cytoxan or with severe respiratory infections should be given Bactrim to protect against Pneumocystis Carinii
What are the protein targets associated with c-ANCA and p-ANCA?
c-ANCA: proteinase 3; p-ANCA: myeloperoxidase
What clinical manifestations besides lung/renal are associated with Wegener’s?
Weight loss, fatigue, fever, proptosis (retro-orbital pseudotumor), palpable purpura, ulcers, subcutaneous nodules, vesicles, joint/muscle pains, mononeuritis multiplex (not as common), pericarditis – positive c-ANCA