LT28-32
104 important questions on LT28-32
What kind of treatment for tumors do you have?
- type of tumor
- growth velocity
- metastases
Palliative treatment
- pain management
- supportive care
Pharmacological therapy
- cytostatics
- hormones
- immunomodulators
- target therapy
What is the difference in growth in normal cells and in tumor cells?
Tumor cells: growth out of control, transcription of proteins that stimulate cell division is high, transcription of proteins that inhibit cell division is low
What are cytostatic drugs?
Alkalyting cytostatics.
Antimetabolites
Antimitotics
Topo-isomerase inhibotrs
- Higher grades + faster learning
- Never study anything twice
- 100% sure, 100% understanding
What are general principles of cytostatic drugs?
No specific inhibitory effects on invasiveness, loss of differentiation or tendency to metastases
Induce damage to DNA synthesis and initiate apoptosis
Main target is cell division, they affect all rapidly dividing normal tissues.
They have toxic effects.
What are general toxic effects of chemotherapy?
Impaired wound healing
Loss of hair
Damage to gastro-intestinal epithelium -> is also a rapid dividing cell
Depression of growth in children
Sterility
Teratogenicity
Nausea and vomiting
What patient characteristics are imporant in cancer?
What treatment is often given in women with breast cancer ER or PR positive?
In elder women the treatment is often tolerated, because they hae less estrogen.
Most of those patients have a very long stabilised disease with this therapy.
No surgery and radiation given.
What is the performance scale?
1: restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature
2: ambulatory and capable of all self-care but unable to carry out any work activites. Up and about more than 50% of waking hours.
3: capable of only limited self-care, confined to bed or chair more than 50% of waking hours
4: completely disables, cannot carry on any self-care. totally confined to bed or chair
5: death
What is the natural course of sarcoma?
What is the natural course of pancreatic cancer?
What is the natural course of small cell lung cancer?
How is cancer diagnosis and staging done?
Supported by imaging (staging): X-rays, ultrasound, endoscopies, MRI, CT, PETscan
Sometimes laboratory investgations.
What is a PET scan?
The radiolabeled sugars you see in the PET scan.
But some cancers don't take those sugars, those are pre-malignant tumors, or tumors who don't have a great potency
What ways does cancer have to spread?
lymphogenic (Nstage)
hematogenic (m stage)
Why do we stage cancer?
Helps estimating prognosis
Helps identifiying eligibility for clinical trials/studies
Makes comparison possible between institutes
Communication
When do you do cancer staging?
- in cancer types with known high prevalence of early metastatic disease (eg lung cancer)
- in cancer that is advanced at presentation N++ breastcancer
If finding metastatic disease has large clinical consequecenes
- refraining from surgery if metastses are found
- adding chemotherapy
NOt every patient with a cancer diagnosis is fully staged
- T1N0 breast cancer: only mammography and ultrasound of axilla, not other imaging
What additional information do you want apart from tissue type and grade?
Growth factors
Gene expression profile: mamaprint
mutations
Lymphovascular space invsasion
Which primary treatment options are there in cancer?
Dependent on stage of disease
Surgery, chemotherapy, radiotherapy, combination
What do you ask yourself when you're considering adjuvant treatment?
how large is the risk for development of metastases? systemic therapy
Why should you give someon with breast cancer adjuvant chemotherapy?
Why should you vie someone with oesophageal cancer adjuvant treatment?
Adjuvnt chemotherapy only treats micrometastatic disease
What questions with: expected disease course after primary treatment?
Should you expect metastases? if so, treatment options?
Should you expect treatment complications?
Does this patient need help or extra guidance?
How are you going to organize the follow-up
Why do we do a follow-up in breast cancer?
After primary treatment the patient can develop distant metastases or recurrens
What differences are there in cancer?
Between types of cancer: breast cancer vs lung cancer, colon cancer vs rectal cancer, SCLC vs NSCLC
What is radiation oncology?
using ionizing radiation to treat cancer
curative intent in many patients
netherlands worldswide leader in research
What is the difference between radiologist and radiotherapist?
As much vs as little radiation as possible
Not the same as nuclear medicine
But with soluble radionucleitides vs linear accellators and fixed sourcess
What is the contribution of radiotherapy to cancer care?
Increase 3% per year due to aging and population increase
50% iraadiated: curative intent/palliative intent
Increasing numbers of long survivors
More awareness of side effects
What is the mechanism of action in radiation/
Repair processes before cell division: suspension of division
Normal cells recognize and repair most of the damge
Cancer cells have less regenerative capacity and die at cell division
Side effects determined by slow down of cell division in surrounding healthy tissue (acute vs late)
Acute side effects: fast growing tissues
Radiotherapy in historical perspective?
1910: radioactivity
1950: cobalt-60
1970: linear accelerators
now: planning treatment, 3DCT, intesnity modulated Rt, image guided RT, brachytherapy, stereotactic RT
2018: proton therapy
With what in the electromagnetic spectrum do we treat patients?
How is external delivery of radiation, how is internal delivery of radiation?
Internal: brachytherapy
Tell the differences between alpha, beta and gamma particles.
Beta particle: high energy stopped later
Gamma: high penetration, only thick layer of concrete could stop it
What kind of radiation could we use for superficial tumors?
What is photon radiation?
What is external beam radiotherapy?
Linear accelerator: on- off
The patient is not radioactive after the treatmetn
What is the mechanism of action of ionizing radiation?
and indirect action: via oxygen radicals
causes double strand DNA damage, single strand DNA damage, losing of peptides
What is the aim of radiotherapy?
higher dose does not always give more tumor control.
Local recurrence free survival, overall survival is comparable
What is the therapeutic window?
Depends on the normal tissue whether you're going to accept the toxicity or not
How can you increase the tumor control?
You have to incresae the cose. But the dose in the normal tissue has to be at the same level, so you get an increased therapeutic window.
So you take measures to change the tolerance boundary.
Daardoor vergroot je je therapeutic window.
How can you increase the therapeutic window?
You can increase the dose, the curve will go to the left
Lower the dose to normal tissue (decreased toxicity)
- shield healthy tissue
- irradiate from multiple directions (treatment planning)
- image guided therapy
Fractination of the total dose: normal tissue can have more dose.
Enhancement sensitivity
What are early side effects of radiotherapy?
swallowing problems, passage problems, nausea, diarrhea, dermatitis, pneumonitis, alopecia
What are late side effects of radiotherapy?
myelopathy
secundary tumors, mutation in normal cell and normall needs to develop to a tumor cells, this takes some years, mostly you find it in the low dose of the radiation field
What are chronic side effects of radiotherapy?
What damage is done in the bone marrow with dose of 1-5% harm (Gy)?
What damage is done in the lung with dose of 1-5% harm (Gy)?
What damage is done in the spinal cord with dose of 1-5% harm (Gy)?
How do you choose fractination schedules?
The fraction of dose varies
The total dose varies
Not all tumor cells are similar in radiation sensitivty
What technical options are there to lower the dose to normal tissue?
Decrease the dose if you give it from 4 fields.
Shielding with led, you can reduce the dose in normal tissue.
Brachtherapy: dose is more locally
What is delineation of treatment volume?
Because you know where the organs are, you know better how to avoid them.
You make daily aligment on tattoos, so that the patientw ill always be in the same positiion
What is clinical target volume? What is planning target volume?
Planning target volume: volume we take into account for movement during therapy.
Lung tumors: you have to get a larger planning target volume than a tumor in the arm which is fixated
What are sollution to account for movements?
- Smaller margings: regional misses?
- image guided radiotherapy IGRT
How can you enhance the sensitivity of tumor cells to radiation?
- chemoradiotherapy
- biological agents (cetuximab in head and neck cancer)
- oxygen to tumor -> better tumor control in radiation
- hyperthermia -> rise of the body temperature to 40 degrees, most of the proteins will be degraded, radiotherapy there is an ehancement of the effect
What is the mechanism of action of chemoradiation?
Different modes of action for radiation and chemotherapy
Normal cells ahve greater variety of escape mechanism and repair the damage
Chemotherapy as radiosensitizer enhances the effect of radiotherapy
Compraed to radiotherapy more toxicity
- synergy in damage inflicted: cisplating adducts to DNA in combination with radiation induces a single strand break, which is more difficult to repair.
- inhibition of post-irradiation repair: cheotherapy can inhibit metabolism
- effects of radiation and chemotherapy in different phases of the cell cycle
- chemotherapy treats hypoxic cells (less radiosensitive)
What are the effects of chemoradiation?
systemic effect chemotherapy, not in all tumors. Sometimes higher dose of chemotherapy and than there is a systemic effect, but no in all tumor types
What are examples of chemoradiation in primary treatment?
What are examples of chemoradiation in adjuvant treatment?
lung cancer
What are examples of chemoradiation in neoadjuvant treatment
rectal cancer
What toxicities in chemoradiation? Acute/Late
Late: function loss (incontinence, swallowing), fibrosis/stenosis, oedema, infertility
In what cancer is curative ressection not possible?
cervical carcinoma
vulvar cancer
esophageal cancer
lung cance rstage III disease, lymph nodes involved in mediastinum, you have to give chemoradiation
What kind of chemotherapy do you have in chemoradiation therapy (CRT)
cisplating/carboplatin -> squamous cell carcinoma
paclitaxel
mitomycin
temozolmide
capecitabine -> rectal cancer
What kind of adenocarcinomas are treated with chemoradiation therapy?
esophageal cancer
cervical cancer
What is the cross effect on distant metastases of chemoradiation thearpy?
- effective treatment of primary tumor helps, mechanism unknown
- direct systemic effect of chemotherapy
- tumor regression predicts survival
- active tumor the prognosis is low, if there was a low tumor response and no tumor could be found, tahn there is a good survivl
- You only find it after surgery
- has become the standard treatment, neo-adjuvant: radiation
What is proton therapy?
One proton enters the body and gives its energy in a certain moment Bragg peak.
After bragg pek no radiation any more int he tissue.
If you give a lot of Bragg peaks you can cover the tumor
What are advantages of proton therapy?
rather spring normal tissue: less side effects, induction of secondary tumor is lower
What are disadvantges of proton therapy?
REstricted in radiation possibilities, you can irradiate photons from every angle, but with protons there is a limit
What are indiciations for proton therapy?
If you give photon therapy , the organs in front will be targeted as well.
Skull base chordoma/chondrosarcoma
Genomically targeted therapy
Chemotherapy
Immune checkpoint therapy
Combination with genoically targeted agent and immune checkpoint therapy.
Which gives the best percent survival?
Immune checkpoint therapy
Genomically targeted therapy
Chemotherapy
But patients will still develop resistance or recurrence.
What is the pahtway of genomics in personalized medicine?
Imaging
Pathology
Clinical chemistry
Genomics
What is DNA next generation sequencing? (NGS)
You use probes, you only sequence the exon regions.
In whole genome seuqncing you sequence 3 billion base pairs and do that several times.
In target sequencing you only sequence the genes.
Probes that are homologous for your genes, these probes can be RNA, that have some markers.
When they bind your target of interset, you digest your probes away and you only sequence the DNA you capture.
What are the steps of next generation sequencing?
2) extract and fragment DNA
3) capture exome DNA with primers
4) recover only exome DNA fragments
5) sequence on next-generation platform
What is SNP testing?
Some polymorphisms are important, see the differences
What are investigational tests?
RNA sequencing
Methylome
What are established tests?
Somatic mutations analyses (targeted gene panel ampliseq cancer hotspot panel V2)
SNPs in genes involved in drug metabolism (pharmacogenomics)
Gene expression panels (oncotype DX/mammaprint for breast cancer)
What is class comparison?
Interregate the genes that are differnetially exprssed between these two groups.
So you compare the classes
What is class prediction?
Results in a test with diagnostic relevance.
If some genes are highly expressed, these patients will respond well to certain therapies.
When you get a patient, you analyze the patient and check whether its profile matches a responder or non-responder.
You predit where this profile fits.
What is class discovery?
You pick heterogenous group of tumors.
You look to the expression profiles and cluster it based on affinity of expression of different genes. You make clusters and try to extract biolgoical or clincial sginificance.
What are challenges in precision medicine?
Result interpretation and explanation: increased complexity of the results that are obtaining is difficult to deal with
Data that you get of patients is very complex.
omputationally intensive: data storage capacity, cloud computing, data transfer speeds
Privacy concers: data security is essential (separate servers for outside facing apps and databases)
What is precision medicine?
What medicine if you have a ER positive tumor
What drug f you have a HER2 receptor?
Which FDA-approved genomic tests are now routine for breast cancer?
Patients falling within the low recurrence score group do NOT need chemotherapy
What are umbrella trials?
Umbrella trial on colorectal cancer and there will be a group with MMR-D, MMR-P, KRAS mutant, KRAS wildtype
1 type of cancer
different genetic mutations
What are basket trials?
multiple types of cancer
1 common genetic mutation
What is cisplatin? And what are the side effects?
Central platinum atom surrounded by two chlorine atoms and ammonia groups
After entering the cell --> reactive comlex
Common side effects:
- severe vomiting and nasea
- myelosuppression is relatively low
- nephrotoxic
Hyperhydration and dialysis prevent nephrotoxicity
In which phase anti-microtubule drugs?
What are deficiencies in growth conditions in G1 phase?
Anti-proliferative factors
DNA damage
Ionising radition
UV
What drugs in replication faults?
DNA damage
Phase s
What drugs in topoII inhibitors?
Phase G2
What is the mechanism of action in antimetabolites?
- Compete with natrural substrate for thea ctive site of an enzyme
- Mimic action of normal metabolites in folic acid cycle and the metabolic pathway of purine and pyrimidine synthesis
- Act during S-phase
What is 5 fluorouracil (5FU)
Common side effects:
Mucositis
Myelosuppresion
Diarrhea
What do mitosis inhibitors?
Mechanism of action:
- binding to microtubules
- stabilize the microtubules by inhibiting their polymerization
- induce cell shape changes
this inhibits cell division
What are topoisomerase inhibitors?
Topoisomerase enzymes: unwind, cut and ligate DNA, crucial role in competition of mitosis + DNA replication.
Topoisomerase inhibitors block tese enzymes and prevent the cell from replication.
- inhibition of topo-isomerase
- inhibition of unwindg, cutting and ligation of DNA
- inhibition of DNA replication and RNA transcription
What are limitations of cytostatic drugs?
Problems with efficacy of chemotherapeutic agents -> resitance to cytostatics
Cytostatics predominantly affect rapidly dividing cells
Do not specificially target cancer cells
They only influence a cells ability to divide and have little effect on other aspect of tumor progression
Cytostatics are associated with high incidence of adverse side effects: bone marrow suppression, alopedcia, mucositis, nausea, vomiting
What kind of resistance to cytostatics can occur?
Requierd: developing during treatment (adaptation or mutation of tumor cells)
What are possible explanation of development of resistance to cytostatics?
-Increased concentration of the target enzyme due to a compensation mechanism (eg methotrexate), could first be very effective, but due to an increase it can be less effective
-Rapid repair of drug-induced lesions (eg alkylating agents)
-Altered activity of target such as topoisoerase II (eg doxorubicin)
-Mutation of various genes
What are side effects of cytotoxic drugs?
Acute emesis: 1-2 h after onset of chemotherapy and can last for 8-24 h
Delayed emesis: usually 24-72 h or later after the onset of chemotherapy
What are examples of 5-HT3 receptor antagonists?
granisetron
What are examples of D2-antagonist
What is target cell therapy?
Make the tumor cell recognizable for immune cell attack.
If you're able to block the receptor, you're able to reduce the cell growth, so you reduce the expression of grow stimulating proteins
What is the function of the HER2 receptor?
this gives growth factor production and proliferation
What are tyrosine kinase inhibitors, name an example.
Imatinib: inhibitor of protein tyorisine kinase, inhibition of cell proliferation, induces apoptosis
What is palliative treatment?
Improves quality of life
What are palliative beneftis of radiotherapy?
reduction of headache and vomiting in raised intracranial presure from CNS metastases
relief of obstruction of bronchus, oesphagus, ureter and lyphatic
preservation of skeletal integrity from metastases in weight-bearing bones
reversal of neurological impairment from spinal cord or optic nerve compression by metastases
What are acute side effects of radiotherapy?
mcuositis, oesophagitis, diarrhoa
alopecia
myelosuppresion
late side effects
secondary malignancies
The question on the page originate from the summary of the following study material:
- A unique study and practice tool
- Never study anything twice again
- Get the grades you hope for
- 100% sure, 100% understanding