Quiz 6 Flashcards
non-target drug delivery
most conventional dosage form - eventually reaches the site of action by distribution and passive diffusion
targeted therapies
reach the right site at the right dose at the right time for the correct duration - pick out the cancer cells
how targeted therapy is different from chemotherapy
uses drugs, targets the cancers specific genes, proteins, or the tissue environment that contributes to cancer growth and survival. takes advantage of the difference between normal cells and cancer cells. sometimes they are used alone, but most often used in combination therapy
targeted therapy can work to
block or turn off chemical signals that trigger growth and division
change proteins inside the cancer cells
stop making new blood vessels that provide nutrition
trigger your immune system to attack cancer cells
carry toxins to the cancer cells to kill specific cells
targeted therapy strategies (4)
- monoclonal antibodies
- antibody directed enzyme prodrug therapy (ADEPT)
- immunotherapy (cellular immunotherapy, virotherapy/oncolytic virus therapy, MAbs)
- gene therapy
delivery carrier strategies
- vectors
- liposomes
- nanoparticles
monoclonal antibodies
the formation of a monoclonal antibody (mAb) is one of the most powerful techniques for incorporating a drug into a site-specific system- mABs are highly specific and recognize only one antigenic determinant or receptor site
naked mab
add nothing to mAB - trastuzumab - cause other tcells/bcells to get rid of what is attached - targets the extracellular domain of the HER2 protein
conjugated MABs
circulates until it finds its target antigen, once bound- releases a toxic agent. thus, less damage to normal cells
radiolabeled MAbs
goes to the side of action and kills, antibody against CD20, antigen on >20% B-cell lymphomas, target B cells if they have CD20 on their surface, non-hodgkins lymphoma, targets only what you want to kill
Ibritumomab tiuxetan
radiolabeled mAbs
trastuzumab
naked mab
chemolabeled MAbs ADC
antibody drug conjugates (ADC), mAb to CD30 attached to chemo drug MMAE, antigen on lymphocytes, hodgkin lymphoma - has a linker that is a synthetic protease-cleavable linker that covalently attaches MMAE to the CD30 directed antibody and releases the agent in the cell -MMAE binds to tubulin disrupting the microtubule network and arresting the cell cycle resulting in apoptosis of the cancer cell
drawbacks to chemolabeled MAbs
- off target toxicity- takes up normal cell if there is not a toxic cells (bistandard effect- once cell dies leads to kill of other cells nearby)
- mAb: linker needs enzymatic reaction, sometimes linker is weak and breaks out early causing cytotoxicity
antibody directed enzyme prodrug therapy
mab used to target the side, the enzyme breaks down the prodrug, works around the timor not in the tumor
bystander effect of ADEPT
capability to kill the surrounding non-dividing/non-expressing tumor cells and it amplifies the drug effect
good ADEPT canidates
small molecules with ability to diffuse inot the tumor tissue
targeted immunotherapy
non-specific immunotherapy and specific immunotherapy (cancer vaccines and virotherapy)
T-cell transfer therapy (CAR-T-Cells), immune checkpoint inhibitors (mAbs)
provenge
cancer vaccine, cellular immunotherapy
virotherapy
designed to replicate tumors (not normal tissue) and initiate an immune response to target cancer cells that have metastasized
CAR T-cells - cellular immunotherapy
engineers patients immune cells (T-cells) to treat their cancer
blood is drawn from patients and T-cells separated
viral vector gene coding for a specific antigen is inserted into the T-cells to produce receptors on their service called chimeric antigen receptors (CARs), speciic to a protein on tumor cells, Eg of antigen: Car T cell therapy furthest along in development target an antigen found on B cells called CD19
keytruda (pembrolizumab)
mAb targets PDL-1
gene therapy
fixing a defective gene in the cell by insertion of a functional gene or group of genes - gene control protein expressed, used to treat, cure or prevent disease by changing the expression of a person’s genes or providing a missing protein via delivery of the gene for that protein - effective in getting the right genes into the accurate cells
sucessful gene therapy requires two main components
- therapeutic gene
2. gene deliver system
why has gene therapy not been very successful
delivery vehicle to target genes to desired cells, internalized and reaches nucleus, not enough expression of the delivered gene
delivery carrier strategies - vectors
carrier- a transfection vector - delivers the therapeutic gene to a patients target cells
vectors used on viral and non-viral
viral vectors
carry a modified or foreign gene, most efficient method for gene transfer, infect target cell and transfer the therapeutic gene using their natural biological mechanism, primary requirement is that virus must be unable to replicate and have no lytic (ability to rupture a cell) activity
examples of viral vectors
retroviruses, adeno-associated virus, parvoviruses, herpesviruses, poxviruses
non-viral vectors- Why and what are they made of ?
naked DNA, because viral vectors cause toxicity, immune and inflammatory responses, gene control and tissue targeting (other approaches use liposome complexes and polymer DNA carriers)
liposomes
- alter the pharmacokinetic profile of drugs
- reduce off-target toxicity
- improve the therapeutic index
usually liposomes cannot release drug without interacting with cells
liposome interactions occur in several ways
- endocytosis
- adsorption
- fusion
- lipid exchange
doxil
liposomal doxorubicin
PEGylated liposome carrier loaded with cytotoxic anticancer drug doxorubicin
diffuses across blood vessels into normal tissues and tumors, retained within the blood pool except at sites of increased vascular permeability such as the liver, spleen and tumors - half life 100x longer than free doxorubicin
cardiotoxic risk is 7x lower than free drug
nanoparticles
improve deliver, therapeutic efficacy, patient outcome
Stealth-like features : evade the immune system • Protective layers : prevent the degradation of biologics (e.g. proteins, DNA) • Targeting moieties : improve specificity and tumor accumulation • Membrane-permeation moieties : improve cell uptake • Imaging agents : assess delivery and dosing • Endosome escape mechanisms : longer half-life • Target-dependent assembly or disassembly : control drug release • Microenvironment sensors. eg: pH, proteases : trigger drug release and cellular uptake • Intracellular targeting moieties : direct drugs to specific intracellular compartments
each new functionality elevates
complexity, cost, regulatory barriers
tumor biomarkers
are antigens and help target -
1. folate receptor 2. EGFR 3. HER2