65
NEW DRUG DEVELOPMENT PRESENTED BY- DR. RAJESH. A. KAMTANE, 2 nd YEAR PG DEPARTMENT OF PHARMACOLOGY,

New drug development

Embed Size (px)

Citation preview

Page 1: New drug development

NEW DRUG DEVELOPMENT

PRESENTED BY- DR. RAJESH.

A. KAMTANE, 2nd YEAR PG

DEPARTMENT OF

PHARMACOLOGY,

MIMS, HYDERABAD

Page 2: New drug development

From the synthesis /identification of the molecule to its marketing, a new drug takes at least 10 years and costs 500-1000 million US$.

New drug development is done as per the guidelines laid down by Schedule Y of Drugs and Cosmetics Act (10th amendment)2001, which were amended later in 2005.

Page 3: New drug development

STAGES IN NEW DRUG DEVELOPMENT

1.Synthesis / isolation of compound : (1-2 years)

2. Preclinical studies : (2-4 years)

3. Scrutiny and grant of permission for clinical trials : (3-6 months)

4. Pharmaceutical formulation, standardization of chemical / biological / immuno-assay of the compound : (0.5-1 year)

5. Clinical studies : phase 1, 2, 3 and long term animal toxicity testing : (3-10 years)

6. Review and grant of marketing permission : (0.5-2 years)

7. Post marketing surveillance : (phase 4 studies)

Page 4: New drug development

Is it safe? Does it work?

Does it work in double blind trials?

Page 5: New drug development

Drug development process can be divided into 3 main phases-

1.Drug discovery phase- during which candidate molecules are chosen on the basis of their pharmacological properties.

2.Preclinical phase- during which wide range of animal studies are performed.

3.Clinical trial phase- during which the lead compound is evaluated for safety and efficacy in human volunteers and patients.

Page 6: New drug development

Drug Discovery Phase

Most new drugs are discovered and identified through one of the following approaches-

1.Random Screening-

It is a sort of blind hitting procedure where new chemical entities (natural or synthetic) are subjected to series of pharmacological screening procedures to explore different types of biological activity.

Such tests include studies on animal behaviour, animal models of the human diseases and on isolated tissues.

Page 7: New drug development

Although these approaches are time consuming, expensive and inefficient in providing fruitful results, sometimes these are valuable, since many drugs like morphine, atropine, digitalis, quinidine were discovered in this way.

Cyclosporine, an immunosuppressant drug was also

discovered during routine screening of fungal products.

Page 8: New drug development

Natural Compound Collections

Page 9: New drug development

Natural Compound Library Screening

Page 10: New drug development

2. Serendipity (happy observation, by chance)

•Sometimes a new use is discovered for an old drug or its side effect finds a new therapeutic application.

Examples-

1. Penicillin was discovered in this way, which started the beginning of antibiotic therapy.

2. Lignocaine (local anaesthetic) and phenytoin (antiepileptic) were later used as antiarrhythmics .

3. Methotrexate, an anticancer drug, is also used for psoriasis.

4. Cyclophosphamide and azathioprine (both cytotoxic drugs) are also used to prevent tissue rejection in kidney transplant.

Page 11: New drug development

3. Rational drug designing-

•Two basic strategies are used in rational drug designing---compound-centered approach and target-centered approach.

•Promising agents, through compound-centered approach, could be obtained from natural products. E.g. penicillin from penicillium notatum, paclitaxel (anticancer drug) derived from Pacific yew tree, cyclosporine (immunosuppressive drug) obtained from fungus.

Page 12: New drug development

•The main disadvantage in obtaining lead products from natural source is that these are often complex molecules which are difficult to be synthesized. It is ultimately difficult to synthesize their modified analogues also. Hence lead optimization becomes difficult.

•Compound centered drug designing can be followed for synthetic products also.

•Drugs can be developed from pharmacological data obtained from structure activity relationship of an established drug. E.g.--- many beta blocking drugs available today are based on propranolol structure, --- many ‘triptans’ are based on structure of sumatriptan.

Page 13: New drug development

Target-centered approach---

•Biochemical or molecular targets are used to search for promising compounds.

•Example—it was known that inhibition of angiotensin-converting enzyme (ACE) blocks conversion of angiotensin 1 to angiotensin 2 and hence lowers the blood pressure. Hence it made sense to look for ACE inhibitors (ramipril, lisinopril, etc) or for angiotensin 2 receptor antagonists (losartan, candesartan, telmisartan) as useful antihypertensives.

•With this approach, there is high possibility of getting useful promising agents for lead optimisation.

Page 14: New drug development

4. Designing of a prodrug or an active metabolite as a drug-

•Prodrug- e.g. levodopa used in treatment of parkinsonism.

•Active metabolite- e.g---Paracetamol, an active metabolite of phenacetin, was introduced as a safe analgesic this way. ---Similarly, active metabolite of procainamide, N- acetyl procainamide, is an effective antiarrhythmic drug which does not cause lupus-like syndrome that occurs with procainamide.

Page 15: New drug development

•After the synthesis or isolation of compound, their purity is ascertained by physico-chemical and analytical studies.

•Next comes the stage of LEAD OPTIMISATION where the aim is to identify one or two drug candidates suitable for further investigation.

Page 16: New drug development

•Three to five years may be spent to come to this stage.

•The promising LEAD COMPOUND is then subjected to preclinical evaluation.

•The clinical trials follow only when the results of preclinical evaluations are encouraging.

Page 17: New drug development

PRECLINICAL STUDIES

Page 18: New drug development
Page 19: New drug development

PRECLINICAL EVALUATION PHASE(ANIMAL STUDIES)

•After synthesizing / identifying a prospective compound, it is tested on animals to expose the whole pharmacological profile.

•Experiments are generally performed on a rodent (rat, rabbit, mouse, guinea pig, hamster) and then on a larger animal (cat, donkey, monkey).

•As the evaluation progresses, unfavorable compounds get rejected at each step, so that only a few out of thousands reach the stage when administration to humans is considered.

Page 20: New drug development

Objectives of animal studies are to evaluate---

1.Activity

2.Toxicity

3.Selectivity and Specificity

4.Mechanism of action

5.Drug metabolism

Page 21: New drug development

•The following types of studies are performed---

1.Pharmacodynamic Studies---

•Here actions relevant to the proposed therapeutic use (and other effects) are studied on animals.

•For e.g., antihypertensive activity of the lead compound on dogs, cats or rats to find out blood pressure changes and other cardiac effects like ECG changes, etc.

Page 22: New drug development

2. Tests on isolated organs, bacterial cultures,etc---performed to detect specific activity, such as antihistaminic, antisecretory, antibacterial, etc.

3. Tests on animal models of human diseases---such as spontaneously (genetically) hypertensive rats, alloxan induced diabetes in rat or dog, etc.

4. General observational test---The drug is injected in tripling doses to small groups of mice which are observed for overt effects. Preliminary clues are drawn from the profile of effects observed.

Page 23: New drug development

5. Confirmatory tests and analogous activities---Compounds found active are taken up for detailed study. Other related activities. e.g antipyretic and anti-inflammatory activity in an analgesic are tested.

6. Mechanism of action---attempts are made to find out the mechanism of action, e.g. whether an antihypertensive is an alpha blocker or beta blocker, etc.

Page 24: New drug development

7. Systemic pharmacology---irrespective of the primary action of drug, its effects on major organ systems such as CNS, CVS, RS, GIT are worked out.

8. Quantitative tests---the dose-response relationship, maximal effect and comparative efficacy with existing drug is ascertained.

Page 25: New drug development

9. Toxicological studies---•Aim – to determine safety of the compound in at least 2 animal species, mostly mouse/rat and dog by oral and parenteral route.

•Types of toxicity studies are-

(1) Acute toxicity- Aim is to find out the acute dose that is lethal to 50% of the animals (LD 50).

Organ toxicity is examined byhistopathology on all animals.

Page 26: New drug development

2. Subacute toxicity-

Aim is to identify the target organs susceptible to drug toxicity.

The animals are maintained at the maximum tolerated doses for a period of 1-3 months so as to allow development of pathological changes.

Finally, animals are killed and subjected to histopathological examination.

Page 27: New drug development

3. Chronic toxicity-

Such studies are important if the drug is intended for chronic use in human beings.

The duration of study may range from one to two years.

These studies may also run simultaneously with clinical trials, to cut short the time factor.

Page 28: New drug development

10. Pharmacokinetic studies-Done after toxicological studies.Information is obtained for its pharmacokinetic parameters (ADME, Vd, BA, t1/2)

11. Special long term toxicity-these tests are generally performed only on drugs which cross phase 1 clinical trials.

12. Reproduction and teratogenecity-effects on spermatogenesis, ovulation, fertility and developing foetus are studied.

Page 29: New drug development

13. Mutagenecity-ability of the drug to induce genetic damage is assessed in bacteria (Ames test), mammalian cell cultures and in intact rodents.

14. Carcinogenecity-drug is given for long term, even the whole life of the animal and they are watched for development of tumours.

Standardizied procedures under ‘Good Laboratory Practices’ (GLP) are laid down for conduct of animal experiments, especially toxicity studies.

Page 30: New drug development

Assessment of Safety Index-

Therapeutic index

Maximum Tolerated Dose (MTD),

No Observable Adverse Effects Level (NOAEL),

No Observable Effects Level (NOEL) and

Human Equivalent Dose (HED) are determined in species similar to humans (like monkeys), finally to calculate First in Human Dose (FIH) which will be latter used in phase 1 clinical trials.

FIH is 1/5 or 1/10th of HED.

Page 31: New drug development

CLINICAL TRIALS

Page 32: New drug development

Ethics Committees

• The ethics committee reviews a protocol before the study is allowed to start. Their job is to ensure that the risks of being in the study are not greater than the potential benefit.

Page 33: New drug development

IRB( Institutional Review Board) IEC (Independent Ethical Committee)

• To ensure the rights and welfare of the participants.

• FDA regulations mandates to review the clinical trial protocols for ethical and legal issues.

Page 34: New drug development

34

IRB

The investigator must furnish the IRB with the following documents for review and approval:

• Trial Protocol• Written Informed Consent Forms• Written Information for Subjects (Advertisements)• Information about compensation to patients• Investigator Brochure• Available (or additional) Safety Information• Investigator’s CV• All amendments to study protocol

Page 35: New drug development

35

IRB

The IRB’s possible responses:• approval or favorable opinion• modifications required for approval• disapproval or negative opinion• withdrawal or suspension of an earlier approval

No subjects should be enrolled until the IRB has issued an approval (21 CFR §56.109)

Page 36: New drug development

Informed Consent

• Eight basic elements of informed consent (21 CFR § 50.25)

1. Trial involves research, purpose of the research

2. A description of any reasonably foreseeable risks or discomforts

3. A description of any benefits to the subject which may reasonable be expected from the research

4. A disclosure of appropriate alternative procedures or treatment that may be available to the subject

Page 37: New drug development

37

Informed Consent

5. A statement describing the extent to which confidentiality of records identifying the subject will be maintained.

6. An explanation as to whether any compensation and whether any medical treatments are available if injury occurs.

7. An explanation of whom to contact for answers to questions about the research and research subjects’ rights .

8. A statement that participation is voluntary

Page 38: New drug development

Clinical Trials & Research 38

Informed Consent

• Participation in clinical trials is always voluntary.

No, thank you, I’d rather not participate.

Yes, I would like to

participate.

Page 39: New drug development

Clinical Trials & Research 39

Informed Consent• Purpose

• Medicine to be studied

• Procedures and schedule

• Risks

• Potential benefits

• Alternatives to participation

• Confidentiality

Page 40: New drug development

Clinical Trials & Research 40

What is a Clinical Trial?Identify a health question.

Develop a plan.

Enroll volunteers and follow the plan.

Study the information collected.

Share the results with others.

Improve treatment.

Page 41: New drug development
Page 42: New drug development

Objectives of a clinical trial are to establish---

1.Safety

2.Efficacy

3.Therapeutic conformation in a large population.

4.Drug metabolism in humans.

5.Unpredicted adverse reactions and new therapeutic applications during wide use in the community.

Page 43: New drug development

CLINICAL TRIALS-

•When a compound deserving trial in man is identified by animal studies, the regulatory authorities are approached who on satisfaction issue an ‘investigational new drug’ (IND) license.

•The drug is formulated into a suitable dosage form and clinical trials are conducted in a phased manner.

Page 44: New drug development

•Standards for design, ethics, conduct, monitoring, auditing, recording and analyzing data and reporting of clinical trials have been laid down in the form of ‘Good Clinical Practice’ (GCP) guidelines by an International Conference on Harmonization (ICH) and Declaration of Helsinki.

NAZI HUMAN EXPERIMENTS

Page 45: New drug development

Declaration of Helsinki-1964

1.The clinical trial must minimize the risk for participants.

2.Provision for care of the patients.

3.Terminate the trial when the risk becomes incompatible with the goals of the trial.

4.Adverse events to be reported immediately to an ethical committee

Page 46: New drug development

Clinical Trials & Research 46

Phases of Clinical Trials

A A A

A

A A A

A

A A A

A

A A A

A

A A A

A

A A A

A

A A A

A

A A A

A

A A A

A

Page 47: New drug development

Phase 0 Clinical trials

Known asHuman Micro dosing.

To confirm whether the drug behaves in human subject as was expected from pre clinical studies.

Page 48: New drug development

Phase 0 Clinical trials

Single sub therapeutic doses of the study drug is given to a small number of subjects ( 10 to 15 ).

Purpose is to gather preliminary data on best PK and PD parameters in humans to take forward for further development.

It gives no data on Safety or Efficacy.

Page 49: New drug development

The clinical studies are conventionally divided into 4 phases-

Phase 1: Human pharmacology and Safety.The objectives of this phase are---

1.To check for safety and tolerability .

2.To determine the pharmacokinetics of the drug in humans.

Page 50: New drug development

3.To determine a safe clinical dosage range in humans.

The common rule is to begin with 1/5th or 1/10th of

the maximum tolerated dose (mg/kg) in animals

and calculating it for an average human body weight

of 70 kg. The drug is then given in small increments till

the therapeutically effective dose is attained by clinical

observation.

Page 51: New drug development

Phase 1 trial is carried out by qualified clinical pharmacologists/ trained physicians in a setting where all emergency/ resuscitative facilities are available.

Page 52: New drug development

Subjects (mostly healthy volunteers, sometimes patients) are exposed to the drug one by one (total 20-40 subjects) , starting with lowest estimated dose and increasing stepwise to achieve the effective dose.

No blinding is done: the study is open label. i.e. both, the investigator and the subjects know what is being given.

Page 53: New drug development

Phase 2: Therapeutic exploration and dose ranging-

•In this phase, the drug is studied for the first time in patients with target disease, to determine its efficacy.

•The main purpose is to decide an end point.

e.g. •pain relief is the end point for testing an analgesic. •reduction in tumour size for anticancer drugs.

Page 54: New drug development

•The study is generally carried out at 2-4 centers.

•Phase 2 can be single or double blind study.

Page 55: New drug development

•Such studies are mainly conducted in two ways-

a.Parallel design---

One group receives the new drug under trial while other group receives control (established drug) or placebo.

Then results in one group are compared with those in other group.

Page 56: New drug development

b. Crossover design---

Here the new drug is alternated with control (established drug or placebo) in the same patient.

In this method, patient acts as his own control.

This reduces the chances of erroneous results due to individual variation amongst the patients.

Page 57: New drug development

Phase 3: Therapeutic Conformation/ Comparison-

•Generally these are randomized, double blind comparative, controlled clinical trials.

•Conducted on a larger patient population (500-3000) by several physicians at many centers.

•Here new drug is compared with previously established drug or placebo, under standardized conditions.

Page 58: New drug development

•Safety, tolerability, and possible drug interactions are assessed on a wider scale.

•Indications are finalized and guidelines for therapeutic use are formulated.

•A ‘new drug application’ (NDA) is submitted to the licensing authority, who if convinced gives marketing permission, with ‘New Drug Status’

Page 59: New drug development

Launch the drug

Page 60: New drug development

Phase 4Drug is placed in the market and patients are monitored for side effects.

Page 61: New drug development

Phase 4: Postmarketing surveillance-

•In phase 4, data on safety, efficacy and tolerability is collected from practicing physicians.

•Uncommon/ idiosyncratic adverse effects and unsuspected drug interactions are detected at this stage.

•Additional indications may emerge from surveillance data.

•The phase 4 has no fixed duration as it is the surveillance phase.

Page 62: New drug development

•During the ‘New Drug Status’, the manufacturer is expected to report any new information about the drug concerning its safety.

•Such Periodic Safety Update Report (PSUR) is to be submitted every six months for first 2 years and then annually for the next 2 years.

•The drug may remain in ‘New Drug Status’ for several years until the Drug Control authorities are confident about its release to unrestricted marketing.

Page 63: New drug development

•Further therapeutic trials involving special groups like children, elderly, pregnant, lactating women, patients with renal/ hepatic diseases, etc (which are generally excluded during clinical trials) may be undertaken at this stage.

Page 64: New drug development
Page 65: New drug development