Cannabis in Acute Migraine Treatment Project - NIH Application 1998

Principal Investigator: Ethan Russo, M.D.


[posted March 1999 on MAPS website]

DESCRIPTION
State the application's broad, long-term objectives and specific aims, making reference to the health relatedness of the project. Describe concisely the research design and methods for achieving these goals. Avoid summaries of past accomplishments and the use of the first person. This description is meant to serve as a succinct and accurate description of the proposed work when separated from the application. If the application is funded, this description, as is, will become public information. Therefore, do not include proprietary/confidential information. DO NOT EXCEED THE SPACE PROVIDED.

Rationale
Cannabis, or marijuana, has been used for centuries for both symptomatic and prophylactic treatment of migraine. It was part of the Western pharmacopoeia for this indication even into the mid-twentieth century. Current anecdotal studies continue to refer to its efficacy for this malady, while biochemical studies of THC and anandamide have provided a scientific basis for such treatment.

Design
Forty adult patients meeting International Headache Society (IHS) criteria of acute migraine with or without aura, with headache frequency of three or more attacks per month, will be recruited. Exclusion criteria will include concomitant use of MAO inhibitor drugs, pregnancy, cardiac conditions, history of drug or analgesic dependency, or affliction with the chronic daily headache variant. A double-blind crossover study design will be pursued. All patients will undergo a detailed screening neurological examination.

After suitable informed consent, study patients will be randomized to one of two groups. Group A patients will initially receive study medications consisting of Marinol (dronabinol, synthetic THC) 10 mg. p.o. plus smoke a placebo marijuana cigarette, or alternatively receive sumatriptan 6 mg. s.c. Group B patients will initially receive an oral placebo capsule resembling Marinol plus smoke a 4% THC content marijuana cigarette, or alternatively, sumatriptan 6 mg. s.c. The pyrolysed Cannabis dose will be titrated to the patients' responses. All patents will be monitored for one hour, at which time they will complete questionnaires regarding symptom-relief employing visual analogue scales. A Folstein Mini-Mental State Examination will also be performed. Blood samples for THC will be drawn at ten minutes and two hours except in those patients who received sumatriptan. Folstein tests and questionnaires will be repeated at the two, three and four hour-hour marks. At the end of three hours, any patient who desires additional symptomatic relief will be offered 1000 mg. of magnesium sulfate IV as a rescue medication. After four hours, patients will be allowed to return home, with a designated driver, or via arranged transportation.

All patients will subsequently complete questionnaires at the twenty-four hour mark to determine efficacy of their treatment with respect to pain levels, nausea, photophobia, and their perceived ability to engage in work or study activities.

Patients will be treated for up to 10 events per three-month block. After a two-week "washout period," groups A and B will crossover regimens. Other parameters of interest in the study will pertain to any rebound headache, the observed study medication side-effect profiles, including the patient's relative ability to function normally post treatment, and subsequent frequency of attacks. Results will be subjected to standard statistical analyses.

PERFORMANCE SITE(S) (organization, city, state)
Department of Neurosciences
Western Montana Clinic
515 West Front St.
Missoula, MT 59807

KEY PERSONNEL. See instructions on Page 11. Use continuation pages as needed to provide the required information in the format shown below.

Name Organization Role on Project
Ethan Russo, M.D. Western Montana Clinic Principal Investigator
Jeannine Mielke University of Montana Research Assistant
Laura Taylor Painter University of Montana Research Assistant
Stuart Hall, Ph.D. University of Montana Consultant
Brian Steele, Ph.D. University of Montana Consultant

PHS 398 (Rev. 5/95)
Page 2BB
Number pages consecutively at the bottom throughout the application. Do not use suffixes such as 3a, 3b.


RESEARCH GRANT

TABLE OF CONTENTS

Page Numbers

Face Page 1
Description, Performance Sites, and Personnel 2
Table of Contents 3
Detailed Budget 4
Budget for Entire Proposed Period of Support 5
Budgets Pertaining to Consortium/Contractual Arrangements 6
Biographical Sketch -- Principal Investigator/Program Director 11
Other Biographical Sketches (Not to exceed two pages for each) 13
Other Support 21
Resources 22
Introduction to Revised Application 23
Introduction to Supplemental Application (Not to exceed 1 page) N/A
a. Specific Aims 24
b. Background and Significance 24
c. Preliminary Studies/Progress Report 42
d. Research Design and Methods 43
e. Human Subjects 48
f. Vertebrate Animals 50
g. Literature Cited 51
h. Consortium/Contractual Arrangements 59
i. Consultants 59
Checklist 64
Personnel Report (Competing Continuation only) N/A

*Type density and size must conform to limits provided in Specific Instructions on page 6.

Appendix is included Appendix (Five collated sets. No page numbering necessary for Appendix)

Number of publications and manuscripts accepted or submitted for publication (Not to exceed 10)

Other items (list):

Ethan Russo -- Selected Publications
Russo, E.B., "Headache Treatments by Native Peoples of the Ecuadorian Amazon: A Preliminary Cross-Disciplinary Assessment," Journal of Ethnopharmacology 36:192-206, 1992.

Russo, E.B., "To Know Them as People," chapter in Mariska, J.A. & van den Pol (Eds.), R., Changes of Necessity, Changes of Choice, Brookline Books, Cambridge, MA, 1993.

Russo, E.B., An Ocelot for a Pillow: Researching Headaches, Hallucinogens, and Hunting Magic Among the Machiguenga in Man?, non-fiction book (inedit)

Russo, E.B., The Last Sorcerer: Echoes of the Rainforest, fiction book, accepted for publication, Inner Traditions International, Rochester, VT, 1997.

Russo, E.B., Medora, R., Parker, K.K., and Thompson, C., "Schedule 1 Research Protocol: An Investigation of Psychedelic Plants and Compounds for Activity in Serotonin Receptor Assays for Headache Treatment and Prophylaxis," Bull. of Multidisc. Assoc. for Psyched. Stud. 7(1):4-8, 1997.

Weber, J.T., O'Connor, M-F, Hayakataka, K., Colson, N., Medora, R., Russo, E.B., and Parker, K.K., "Activity of Parthenolide at 5HT2A Receptors," Journal of Natl. Prod. 60:651-653, 1997.

Russo, E.B., "Cannabis for Migraine Treatment: The Once and Future Prescription: An Historical and Scientific Review with Suggestions for Subsequent Studies," Pain 76(1):3-8, 1998.


Studies in Progress
Studies on feverfew, Tanacetum parthenium, and its use as a migraine prophylactic, with Drs. Keith Parker, Rustem Medora, and Chuck Thompson, University of Montana.

Serotonin receptor pharmacology studies on approximately 100 species of medicinal plants collected in Parque Nacional del Manu, Peru, and Belize, with and the above team.

Biochemistry of neotropical gesneriads with reported ethnobotanical uses for headache or psychoactive effects, supported by the American Gloxinia and Gesneriad Society ($1500), with the above team.

Clinical studies on aromatherapy treatment of migraine in process.
Principal Investigator: Russo, Ethan Budd


Personnel
Ethan Russo, M.D. (Principal Investigator): Dr. Russo is a clinical child and adult neurologist at the Western Montana Clinic, Clinical Assistant Professor of Medicine at the University of Washington, Adjunct Associate Professor in the Department of Pharmacy at the University of Montana, and Director of the Inpatient Pain Treatment Program at St. Patrick Hospital, Missoula, Montana. Dr. Russo has had a long research interest in migraine, particularly in the area of ethnobotanical treatments. He is involved in both field-study in the Amazon, and bench research on serotonin receptor activity of potential rainforest plants for this indication.

Dr. Russo has served as an article reviewer for the journal Headache, scientific advisor for Shaman Pharmaceuticals, and medical consultant on herbal treatments for various magazine articles and books. He is former director of the Western Montana Muscular Dystrophy Association Clinic. He was an expert witness in fourteen cases for the Vaccine Injury Compensation program, Department of Health and Human Services, Washington, D.C. Additionally he has served for several years on the Ethics Committee of St Patrick Hospital, Missoula, MT.

He is the author of several papers, two books, one of which is due to be released in 1999.

Dr. Russo will be principal investigator, and oversee day-to-day operation of this project. He will direct publicity and patient recruitment for the study, approve or disapprove prospective study subjects, oversee study drug administration, and troubleshoot patient clinical problems. He will also chair team meetings, supervise data analysis, and preparation of potential publications from the resulting data. Dr. Russo will also administer the grant. A 30% salary support is requested for Dr. Russo for two months of patient recruitment, six and a half months of drug study trials, and one month of data analysis.

Jeannine Mielke (Research Assistant): Ms. Mielke is a doctoral candidate in psychology at the University of Montana, and psychometrician at the Western Montana Clinic. She will be responsible for subject recruitment, confirmation of eligibility, assistance in obtaining informed consent, enrollment of subjects, collection of subject data, data management, statistical generation and preparation of study results. We are requesting 25% salary support for 9 months for Ms. Mielke = $3600.

Laura Taylor Painter (Research Assistant): Ms. Painter is a doctoral candidate in psychology at the University of Montana, and psychometrician at the Western Montana Clinic. She will be responsible for subject recruitment, confirmation of eligibility, assistance in obtaining informed consent, enrollment of subjects, collection of subject data, data management, statistical generation and preparation of study results. We are requesting 25% salary support for 9 months for Ms. Painter = $3600.

Stuart Hall, Ph.D.: Dr. Hall is an Associate Professor in the Department of Psychology at the University of Montana. His area of expertise is clinical neuropsychology and he has extensive training in physiological psychology. He is a member of both the clinical and experimental faculty. Dr. Hall will provide consultation on methodology, clinical measures, data collection, and data interpretation. He will supervise research assistants conducting clinical assessments of subjects and will assist in the preparation of abstracts and manuscripts reporting the results of the study. We are requesting 5% salary support for Dr. Hall.
Salary: $2250.00
Fringe: $ 528.75
TOTAL: $2778.75

Brian Steele, Ph.D.: Brian M. Steele, Ph.D. (Consultant): Dr. Steele is a Visiting Assistant Professor of Mathematics at the University of Montana, Missoula. He is an expert on the statistical analysis of longitudinal studies and related statistical designs. His Ph.D. dissertation developed an improved approach to estimation for generalized linear mixed models, a large class of statistical models that includes the Pilot Study design as a prototype. Recently, he has developed a previously unavailable general approach to hypothesis testing under the generalized linear mixed model. He has been a statistical consultant for the past ten years, working primarily on problems arising in biology and ecology. He has published several papers on design and analysis of longitudinal monitoring studies. Dr. Steele has been a Professor at University of Montana for two years and has been course coordinator of both the Linear Mathematics and Introductory Statistics courses, positions that demand extensive planning and supervision of teaching assistants. Dr. Steele will assist researchers to insure that the study yields statistically valid data. He will be responsible for the statistical analysis and interpretation of the study results and he will be responsible for the statistical content of abstracts and manuscripts reporting the findings of the study. We are requesting 33% salary support for Dr. Steele for 3 months: $4133 + $783 fringe = $4916.

Study Nurses: Two RNs will be hired for this position for a projected two months of patient recruitment, the six and one half months of patient drug trials, and one month of data analysis. They will assist in patient enrollment, administration of study medication, and administration of tests. They will also perform venipunctures for the THC assays, and magnesium sulfate administration. They will monitor study patients, and assess side effects, and assist in follow-up. Two prospective candidates have been interviewed and have expressed interest in the position. 100% salary support is requested for this position for the 9 month period: $16/hr. + 15% fringe = $29,440 x 2 = $58,880.

Consultant Costs
Drs. Steele and Hall, and Ms. Mielke and Ms. Painter will serve as consultants as above.

Equipment
Funds are requested to purchase a computer system for the Study Nurses: $3000 plus software $500 = $3500

D. Travel
Costs associated with travel to one medical meeting are requested for the principal investigator to present study results:

Airfare to New York: $600 (government rate)
Ground transportation: $75
Per diem: $36/ day x 3 days: $108
Hotel: $125/night x 3 nights: $375

Patient Care Costs
Outpatient

Alterations and Renovations
These will be included in a planned renovation by the Western Montana Clinic in 1998. Some minor renovations for ventilation may be needed = $1000.

Other Expenses
Funds are requested to purchase 10 mg. Marinol capsules for 40 subjects. Each might be expected to utilize 5 capsules over the 20 possible trials. Costs would then be 5 capsules/subject x 40 subjects = $2692.

Funds are requested to purchase sumatriptan for the 40 subjects. Each would receive up to 10 injections during the study, incurring costs of: 400 injections x $25/ injection = $10,000.

In the course of the study, the 40 subjects may require up to 5 trials each of Cannabis. Therefore, 200 marijuana cigarettes of 4% THC content are requested.

In the course of the study, the 40 subjects may require up to 5 trials each of placebo Cannabis. Therefore, 200 placebo cigarettes of 0% THC content are requested from NIDA.

200 placebo capsules resembling Marinol are requested from NIDA or the manufacturer: cost unknown.

Costs are requested for an estimated 30% of total patient trials where supplemental magnesium sulfate is needed: 800 trials x 0.3 = 240 doses x $10/ treatment = $2400.

A small number of sumatriptan inhalers may be needed for study patients intolerant of, but not truly allergic to sumatriptan injections.

Clinical Laboratory Costs:
Serum THC assays will be required of patients receiving dronabinol, and smoked Cannabis: 2 assays/session x (200 Cannabis sessions + 200 dronabinol sessions) = 400 x 2 assays @ $100/test = $80,000.

Additionally, venipuncture costs are $4 x 400 x 2 = $3200.

Shipping of samples: $15 x 400 (or less if batched) = $6000.


Other Costs:
Reimbursement for patient sessions is requested @ $30 per session. Costs would thus be 40 subjects x maximum of 20 sessions/subject x $30/session= $24,000.

Reimbursement is requested for taxi service for study subjects who are unable to provide the requested designated driver. Hopefully, this will be required in no more that 25% of instances: 40 subjects x 20 sessions x $10 taxi fare x 2 ways x 0.25 = $4000.

Funds are requested for 2 ventilation fans, and 4 bean bag chairs for patients: 6 x $60/piece = $360.

Funds are requested for room rent from St. Patrick Hospital: $ 225/month x 10 months x 2 rooms = $4500.

Funds are requested for a telephone for the study room: $ 25/month x 10 months = $250.

Funds are requested for postage: $0.32 x 12 mailings/patient x 40 patients + $0.32 x 200 for recruitment to area physicians = $218.

Funds are requested for advertisements in local newspapers to aid in recruitment of study subjects: $200/ad to run 3 occasions = $600

3 alpha-numeric pagers and 3 cellular telephones are requested for study use for 10 months: (3 x $20/mo. x 10) + (3 x $24.44/mo. x 10) = $1332.

Funds are requested for a safe to contain sensitive study materials, such as patient files and study medications = $1000.

Funds are requested for the Investigational
Review Board fee = $500

Funds are requested for a scale to weigh unused portions of marijuana and placebo marijuana cigarettes = $1200.

No indirect costs will be associated with this study, inasmuch as Dr. Russo will administer it.


OTHER SUPPORT

Russo, Ethan

ACTIVE
American Gloxinia and Gesneriad Society 7/96-7/97 $1500

The goal of this grant is to study serotonin receptor activity of neotropical gesneriads with reported psychoactive effects or use in headache treatment.

Overlap: 0%

Multidisciplinary Association for Psychedelic Studies
Fall '96-1997 $2500
The purpose of this grant is to study serotonin receptor activities of psychoactive plants and compounds that may have utility in migraine treatment.

Overlap: 0%

3) Multidisciplinary Association for Psychedelic Studies
3/97 $3500
1/98 2500
This grant was to aid in the preparation of this NIH proposal.

Overlap: 100%


HALL, S.

ACTIVE
291731 (Hall) 4/8/96 - 8/31/96 extended through 8/31/97
MONTS-NSF EPSCoR Program $24,000
The Detection of Malingering Using
Standard Neuropsychological Measures:
The Utility of Response Latency

The major goal of this study is to determine if measuring response latency on two standard neuropsychological tests will significantly contribute to the ability to identify individuals who are malingering on these tests.

OVERLAP
None


Brian Steele: none
Jeannine Mielke: none
Laura Painter: none


RESEARCH PLAN

Introduction to Revised Application:

This application has been substantially revised, and in the case of the study protocol, totally so.
The background section has been supplemented with a great deal of historical and experimental material.
The re-formulated study protocol has been written so as to address criticisms of the 1997 application in the "Notification of Scientific Review Action" (Appendix), and to adhere to recommendations of the Headache Classification Committee of the International Headache Society (1988: references and Appendix) with respect to study design.
Additionally, very close attention was paid to recommendations from the 1997 NIH Workshop on the Medical Utility of Marijuana, and particularly the presentation by Robert Temple, M.D., "Clinical Trial Considerations with Marijuana." Both are included in the Appendix, and the PI would urge reviewers to examine these materials.
Specifically, the proposed study design is now double-blind crossover employing a "double dummy" technique, so as to compare the effects of smoked Cannabis with dronabinol, and injected sumatriptan.
The patient sample has been increased to 40. Patient exclusions have been clarified, as have safeguards with respect to pregnancy, and contraception.
All study subjects will be examined neurologically by Dr. Russo to confirm diagnosis of migraine, and exclude space-occupying lesions, and other causes of headache in the patient population.
Study subjects will be observed for four hours with hourly testing of mental status and completion of questionnaires employing digital analogue scales, much as previously. A provision for rescue medication employing intravenous magnesium sulfate has been added, in the event that study drugs are ineffective after 3 hours.
Increasing to two the number of study nurses, and markedly increasing hours of availability for the performance of study trials will enhance clinical coverage and troubleshooting.
Other minor changes and details are included in the protocol itself.


A. Specific Aims:
The primary goal of this study is to assess the efficacy of smoked Cannabis and oral dronabinol in treating acute migraine headache. This will be examined through a randomized double-blind crossover study comparing these two agents to control placebo Cannabis and control placebo dronabinol capsules vs. injected sumatriptan in a group of chronic migraineurs. In the course of this study, we hope to assess the relative practicality of these modalities as alternative migraine treatments, and monitor attendant side effect profiles.

Hypothesis 1: Pyrolysed Cannabis may ameliorate the pain and nausea associated with migraine.

Hypothesis 2: Oral dronabinol may also show efficacy in this regard, but to a lesser degree, and with a slower response than smoking Cannabis.

A secondary goal is to assess whether periodic use of THC products in the study group alters the frequency or severity of migraine attacks.

Hypothesis 3: THC use may exert a prophylactic effect in migraine.

Rationale: If the current pilot study demonstrates therapeutic efficacy of dronabinol or smoked Cannabis in acute migraine treatment, with attendant safety, additional in-depth, long-range studies may be warranted.

B. Background and Significance

B.1 Historical Data on Use of Cannabis in Migraine Treatment:

Note: An abbreviated version of this discussion is available in: Russo, E.B., "Cannabis for Migraine Treatment: The Once and Future Prescription?: An Historical and Scientific Review," Pain 76(1):3-8, 1998 (Appendix).
Archeological records substantiate an ongoing association between man and Cannabis. It is a member of the plant family, Cannabaceae, with botanical origin in Eastern or Central Asia, possibly in the Pamir Plain (Camp, 1936). The number of species in the genus remains controversial. Some botanists retain all members of the genus as one polymorphic species, while others (Emboden, 1981; Schultes & Hofmann, 1980; Schultes et al., 1974) have exhaustively documented three Cannabis species: sativa, indica, and ruderalis. All specimens contain the psychoactive chemical delta-9-tetrahydrocannabinol (THC) to some degree.
It is claimed that Cannabis use occurred in central Europe by the Bylony culture as much as 7000 years ago (Kabel"k, Kreje" & Santavy, 1960). Hermann Busse uncovered physical evidence for its early use in Europe in 1896 in Wilmersdorff (Brandenburg), Germany in the form of a funerary urn that contained Cannabis sativa leaves and fruit (Busse, 1897). This was subsequently dated to the 5th Century B.C.E. (La Barre, 1980; Andrews & Vinkenoog, 1967).
The first records of Cannabis' use may occur in the P'n-tsao Ching, a traditional Chinese herbal written down in the 1st or 2nd centuries, but said to be based on the oral traditions passed down from the Emperor Sh'n-nung in the third millenium B.C.E. The text noted that the plant fruits "if taken in excess will produce hallucinations (literally "seeing devils")(Li, 1974).
Earlier written documentation of medicinal use in China has also been claimed (Emboden, 1972):

By the fifteenth century B.C. the Chinese book known as the Rh-Ya was compiled, and in it there is mention of the herb Ma, the Cannabis sativa plant. Not only were the fibers and potent resins employed at this time, but the Rh-Ya describes the first ritualistic or shamanistic use of the plant.

The Atharva Veda of India, dated to between 1400 and 2000 B.C.E. referred to a sacred grass, bhang, and medicinal references to Cannabis were cited by Susrata in the sixth to seventh centuries (Chopra & Chopra, 1957). One author (Dwarakanath, 1965), noted Ayurvedic preparations called Rasachandrika vati and Mahalakshmivilasa rasa, said to contain Cannabis, indicated for:

Diseases of the head including neuralgic headaches, haemicrainia etc. (Shiroaroga [term for migraine])

Although many authorities have cited an absence of Cannabis in Ancient Egypt, Nunn (1996) cited no less than five supporting experts that it was employed in fact (Mannische, 1989; Ghalioungui, 1987; Charpentier, 1981; Faulkner, 1962; Dawson, 1934):

There is general agreement with the view of Dawson that shemshemet means cannabis, and the identification was strongly supported by the use of hemp in rope making. As a drug, it has remained in active use ever since pharaonic times. It does not appear very often in the medical papyri, but it was administered by mouth, rectum, vagina, bandaged to the skin, applied to the eyes and by fumigation.

Mannische (1989) also has cited evidence of Cannabis use in Ancient Egypt in the Pyramid Texts of the mid 3rd millenium B.C.E. Physical proof includes discoveries of hemp remnants in the tomb of Akhenaten (Amenophis IV) around 1350 B.C.E., and Cannabis pollen in the tomb of Rameses II, who died in 1224 B.C.E.
The Zend-Avesta, the holy book of Zoroastrianism, which survives only in fragments, dating from around 600 B.C.E. in Persia, alludes to the use of Banga in a medical context, which is identified as hemp by the translator (Darmesteter, 1895).
Campbell documented 29 citations of Cannabis in Assyrian medical papyri, and attested to its analgesic and psychogenic effects by various methods including fumigation (Campbell, 1949).
Longstanding debate has occurred as to the veracity of Cannabis use in the Bible. Benet (1975) proposed such a theory on a strong philological basis (see also: Benetowa, 1936):

Both in the original Hebrew text of the Old Testament and in the Aramaic translation, the word kaneh or keneh is used either alone or linked to the adjective bosm in Hebrew and busma in Aramaic, meaning aromatic. It is cana in Sanskrit, qunnabu in Assyrian, kenab in Persian, kannab in Arabic and kanbun in Chaldean. In Exodus 30:23, God directs Moses to make a holy oil composed of "myrrh, sweet cinnamon, kaneh bosm and kassia." In many ancient languages, including Hebrew, the root kan has a double meaning ñ both hemp and reed.

Current estimates posit the writing of Exodus to between 900-600 B.C.E., while referring to much earlier events. Although the issue of its use in the Bible has been hotly debated, physical evidence of medicinal Cannabis use in Israel/Palestine was recently discovered (Zlas et al., 1993). In a burial tomb in Beit Shemesh (midway between Jerusalem and Tel Aviv), the skeleton of a 14 year old girl was found along with 4th century bronze coins. Contained in her pelvic area was the skeleton of a term fetus, of sufficient size to disallow a successful vaginal delivery. In her abdominal area, gray carbonized material was noted and analyzed, yielding TLC and NMR spectroscopy evidence of delta-6- tetrahydrocannabinol, a stable component of Cannabis. The authors stated:

We assume that the ashes found in the tomb were cannabis, burned in a vessel and administered to the young girl as an inhalant to facilitate the birth process.

They further remarked that Cannabis retained an indication as an aid to parturition into the 19th century, as above noted.
The classical Greek literature also documents knowledge of the inebriating actions of Cannabis. Herodotus, circa 450 B.C.E., described how the Scythian people set up tents, heated stones and threw Cannabis seeds or flowering tops upon them to create a vapor (Herodotus, 1954):

- for when they have parties and sit round a fire, they throw some of it into the flames, and as it burns it smokes like incense, and the smell of it makes them drunk just as wine does us; and they get more and more intoxicated as more fruit is thrown on, until they jump up and start dancing and singing.

Physical evidence of Scythian use of Cannabis has been unearthed (Rudenko, 1970; Artamonov, 1965). The "Frozen Tombs" of Pazyryk, dated to the 5th to 3rd centuries B.C.E., were excavated earlier this century in the Altai Mountains, south of modern-day Novosibirsk near the Mongolian and Chinese borders. Artamonov (1965) describes:

One particularly interesting apparatus was a kind of cone-shaped miniature tent, covered with a felt or leather rug, standing over a copper censer. Hemp seeds found on the spot suggest that this contrivance was a special enclosure that could be filled with narcotic smoke from the burning seeds.

Rudenko (1970) also described a leather flask containing hemp seeds, and asserted:

Here it will be merely remarked that smoking hemp, like smoking hashish, took place without a doubt not just as a ceremony of purification after burial but in ordinary life; hashish was used as a narcotic.

In the 1st century, Diosocorides published his Materia Medica, perhaps the first pharmacopoeia in the Western World. He described the plant and its analgesic role (Dioscorides, 1968):

Cannabis is a plant of much use in this life for ye twistings of very strong ropes, it bears leaves like to the Ash, of a bad scent, long stalks, empty, a round seed, which being eaten of much doth quench geniture, but being juiced when it is green is good for the pains of the ears.

In the 2nd century, the Greek physician Galen expounded on medical indications, mainly gastrointestinal (Brunner, 1977), but also noted of Cannabis (Galen):

If consumed in large amounts, it affects the head by sending to it a warm and toxic vapor.

Medicinal use in the classical world extended in to the early Middle Ages through Paulus Aeginata (Paul of Aegina) in his 7th century Epitome, wherein Cannabis was employed as a carminative (Lewis et al., 1971).
The medicinal use of Cannabis as hashish has been well documented in early Islamic texts (Lozano, 1997). A psychoactive effect was observed by Jabir ibn Hayyan in the Kitab al-Sumum in the 8th century (Lewis et al., 1971). More to the point, Abu Mansur ibn Muffawak ibn Ali al-Harawi in the 10th century described the use of Cannabis fiber for making rope, and the plant to treat headache (Lewis et al., 1971).
Cannabis also figured in the medical writings of Avicenna (ibn Sina) in the 10th century, and those of Maimonides (Moses ben Maimon) in the 12th century (Rosner, 1979).
In 13th century, Umar ibn Yusuf ibn Rasul suggested Cannabis for ear and head pain (Lewis et al., 1971).
European awareness of the psychoactivity of Cannabis was re-kindled with the writings of Garcia da Orta, a 16th century Portuguese visitor to India. In addition to recognizable descriptions of the plant, and a good illustration, the author noted sedative and appetite-stimulating properties in his 1563 book (da Orta, 1913).
More or less contemporaneously, Rabelais wrote of Cannabis in his Gargantua et Pantagruel, including an excellent botanical description of the plant and including medicinal uses (Robinson, 1946).
In 1649 England, Culpeper described various uses of hemp in his famous herbal (Culpeper, undated):

The decoction of the root allays inflammations of the head, or any other parts; the herb or the distilled water of it, does the same.

In 1712, Engelbert Kaempfer published his Amoenitatum Exoticarum Politico- Physico-Medicarum, in which he described the psychogenic effects of Cannabis as employed in Persia and India (Dolan, 1971; Kaempfer, 1996).
The medical use of Cannabis, or what became known as "Indian hemp" was reintroduced to the West in 1839 (O'Shaughnessy, 1839). His treatise on the subject dealt with the apparent utility of a plant extract administered to patients suffering from rabies, cholera, tetanus, and infantile convulsions.
Over the next half-century, Cannabis was recognized as helpful other conditions, including headache. The noted nineteenth century physician, Weir Mitchell (1874) wrote:

It is necessary at times to do something to give immediate relief to the too prolonged pain, and in these cases a combination of cannabis indica and morphia answers very well

Also in 1874, a popular textbook, Practical Therapeutics stated (Waring, 1874):

Of a good extract, gr. 1/4 to gr. ?, rarely gr. j, in the form of pill, is very effective in some forms of neuralgia, particularly -Migraina. Even in the severest and most intractable forms it often palliates greatly. It should be given every night, whether there be pain or not.

These claims support both acute and prophylactic indications of Cannabis for migraine.
In 1887, Dr. Hobart Hare published an article that dealt with this indication in detail (Hare, 1887):

CANNABIS INDICA has been before the profession for many years as a remedy to be used in combating almost all forms of pain, yet, owing to the variations found to exist as to its activity, it has not received the confidence which I think it now deserves. - Within a few years this drug has become particularly prominent in connection with its use in migraine, particularly when used in conjunction with gelsemium [Gelsemium sempervirens (L.) Ait. Loganiaceae, yellow jessamine], although of the two remedies the hemp is by far the most active agent in subduing the pain and preventing other attacks.
-I have certainly seen very severe and intractable cases of migraine successfully treated by this remedy, not only in regard to the attack itself, but by acting as a prophylactic.
-so far no case of fatal poisoning from its ingestion has been recorded as occurring in the human being.
-Cases of migraine treated in this way, when the disease does not depend on any distinct organic lesion, are in a large proportion of instance either entirely cured or greatly benefited, the attacks even when they recur being considerably farther apart.
-The advantages in its use over that of opium consist chiefly in the absence of prostration and nausea after its ingestion, and in the partial lack of soporific power which it possesses as compared to the opiate, for in certain cases sleep is not always desirable when pain is to be removed. That cannabis
indica has, however, marked powers as a soporific is not to be denied. Added to these advantages is the fact of its failure to produce serious symptoms even if very large doses be taken, although I have found the efficient dose of a pure extract of hemp to be as powerful in relieving pain as the corresponding dose of the same preparation of opium.
-During the time that this remarkable drug is relieving pain a very curious psychical condition sometimes manifests itself; namely, that the diminution of the pain seems to be due to its fading away in the distance, so that the pain becomes less and less, just as the pain in a delicate ear would grow less and less as a beaten drum was carried farther and farther out of the range of hearing.

Another British physician agreed with the indication of Cannabis for headache (Fox, 1897):

I understand by migraine a periodical nerve storm ñ- For the relief of the paroxysms antipyrin and phenacetin have often been in my experience successful. ñ- But I am accustomed to rely much upon cannabis indica, having had a pretty large experience of this remedy. The extract, often combined with cascara sagrada [Rhamnus purshianus DC Rhamnaceae], has controlled many, if not most, cases of migraine.

In the same year the following blurb was attributed to "G.O.M." under the heading, "Letter From London (From Our Own Correspondent)" (Mackenzie, 1887):

Dr. Stephen Mackenzie says that in doses of one-half grain Indian hemp, night and morning, and continued some time, is the most valuable remedy he has met with in the treatment of persistent headache.

A doctor in India wrote of Cannabis indica (McConnell, 1888), and how proper storage was key to therapeutic response:

Where care is taken in this respect, the therapeutic value of the drug in certain affections of the nervous system- tetanus, neuralgia, migraine - has been repeatedly recorded by competent observers, and its employment for the relief of such affections is well understood and more or less extensively resorted to.

Dr. William Gowers is now considered one of the founding fathers of modern neurology. For treatment of migraine, he wrote (Gowers, 1888):

Most relief is afforded to the pain by a good dose (thirty or forty grains) of bromide, and its effect is increased by the addition of five or ten minims of tincture of Indian hemp.

For treatment of "headache," he stated:

Sedatives are very uncertain in their influence. Opium and morphia are seldom useful, and often do more harm than good, in consequence of the indirect effect of the constipation that is produced. Gelsemium and Indian hemp frequently lessen the pain, the former chiefly in neuralgic forms about the front of the head, the latter not only in neuralgic, but in anaemic, and also other ill-defined forms of headache.

The next year, a treatise was written on the use of rectal preparations of Cannabis (Farlow, 1889):

Cannabis has few equals in its power over nervous headaches such as women with pelvic troubles are subject to.

The Lancet published an article on Cannabis indica by J. Russell Reynolds, personal physician to Queen Victoria (Russell, 1890) which stated:

Indian hemp, when pure and administered carefully, is one of the most valuable medicines we possess.

In relation to its use in headache, Reynolds said:

Migraine: Very many victims of this malady have for years kept their suffering in abeyance by taking hemp at the moment of threatening, or onset of the attack.

In the following year, the British Medical Journal published a short report, "On the Therapeutic Value of Indian Hemp" (Suckling, 1891), which stated:

In migraine the drug is also of great value; a pill containing º gr. Of the extract with or without a º gr. of phosphide of zinc will often immediately check an attack, and if the pill will be given twice a day continuously the severity and frequency of the attacks are often much diminished. I have met with patients who have been incapacitated for work from the frequency of the attacks, and who have been enabled by the use of Indian hemp to resume their employment.

In A Text-Book of Materia Medica and Therapeutics (Cowperthwaite, 1891), once more, Cannabis indica was indicated for migraine treatment.
The same year, it was written of Cannabis (Mattison, 1891):

-its most important use is in that opprobium of the healing art- migraine.
-Ringer says: "No single drug have I found so useful in migraine." He thinks it acts well in all forms, but seems most useful in preventing rather than arresting. He deems it specially effective in attacks due to fatigue, anxiety, or climacteric change. Dr. E.C. Seguin, in 1877, commended it highly.
Dr. Wharton Sinkler, in a paper on migraine, gives first place to cannabis, and thinks it of more value in this form of headache than any other. Richard Green, who first commended it in this complaint, thinks it not only relieves, but cures; in nearly all cases giving lasting relief.
Anstie commends it in migraine and the pains of chronic chloral and alcohol taking. In his work on neuralgia-the best ever written, and one which I advise every one to read-he says: "From one-quarter to one-half grain of good extract of cannabis, repeated in two hours, if it has not produced sleep, is an excellent remedy in migraine of the young. It is very important in this disease that the habit of long neuralgic paroxysms not be set up."

Drawing subsequently from his own experience, Mattison stated:

Failure with hemp is largely due to inferior preparations
-I close this paper by asking attention to the need of giving hemp in migraine. Were its use limited to this alone, its worth, direct and indirect, would be greater than most imagine. Bare in mind the bane of American women is headache. Recollect that hemp eases pain without disturbing stomach and secretions so often as opium, and that competent men think it not only calmative, but curative.
-Indian hemp is not here lauded as a specific. It will, at times, fail. So do other drugs. But the many cases in which it acts well, entitle it to a large and lasting confidence.
My experience warrants this statement: cannabis indica is, often, a safe and successful anodyne and hypnotic.

Cannabis in its various forms remained the focus of intense scrutiny, and continued to have its critics. Because of concerns of its dangers, the British and colonial authorities in India organized a study of all aspects of the issue (Indian Hemp Drugs Commission, 1893-1894). Its findings after exhaustive investigation and testimony exceeded 3000 pages, and were summarized as follows (Abel, 1980):

(1) Moderate use of cannabis drugs had no appreciable physical effects on the body. As with all drugs, excessive use could weaken the body and render it more susceptible to diseases. Such circumstances were not peculiar to cannabis, however. (2) Moderate use of cannabis drugs had no adverse effect on the brain, except possibly for individuals predisposed to act abnormally. Excessive use, on the other hand, could lead to mental instability and ultimately to insanity in individuals predisposed by heredity to mental disorders. (3) Moderate use of cannabis drugs had no adverse influence on morality. Excessive usage, however, could result in moral degradation. Although in certain rare cases cannabis intoxication could result in violence, such cases were few and far between.

The commission subsequently dissuaded any governmental suppression of usage of Cannabis drugs.
An American 1898 drug handbook stated the following under "Actions and uses" for Cannabis (Lilly, 1898):

Not poisonous according to best authorities, though formerly so regarded. Antispasmodic, analgesic, anesthetic, narcotic, aphrodisiac. Specially recommended in spasmodic and painful affections; for preventing rather than arresting migraine;

In 1900, a brief report on Cannabis indica appeared (Lewis, 1900), which stated, in part:

In migraine, hemicrania, neuralgias, and headache due to eye-strain, it may be used with marked success

A textbook of the era (Marshall, 1905) indicated that Indian hemp had been supplanted by other medicines for some indications, but:

It appears, however, to be useful in headache of a dull continuous character. The extract in the form of pills is usually administered.

As late as 1915, Sir William Osler, the acknowledged father of modern medicine stated of migraine treatment (Osler, 1915):

Cannabis indica is probably the most satisfactory remedy. Seguin recommends a prolonged course of the drug.

This statement once more supports its use for both acute and prophylactic treatment of migraine.
In 1916, in a quotation attributed to Dr. Dixon, Professor of Pharmacology, Kings' College, and the University of Cambridge (Ratnam, 1916), reference is specifically made to the therapeutic effects of smoked Cannabis for headache treatment:

In cases where immediate effect is desired, the drug should be smoked, the fumes being drawn through water. In fits of depression, mental fatigue, nervous headache, feelings of fatigue disappear and the subject is able to continue his work refreshed and soothed.

In 1918, The Dispensatory of the United States of America stated (Remington & Wood, 1918):

Cannabis is used in medicine to relieve pain, to encourage sleep, and to soothe restlessness. -For its analgesic action it is used especially in pains of neuralgic origin, such as migraine, but is occasionally of service in other types.

In the German literature, Cannabis use by extract or smoking was held to be an "outstanding agent" (Dinand, 1921).
In 1922 Hare still advocated use of Cannabis noting, "For the relief of pain, particularly that depending on nerve disturbance, hemp is very valuable." He went on to state (Hare, 1922):

In true migraine with hemianopsia this treatment is often most effectual in aborting the attack. The prevention of further attack is to be attained by the use of smaller amounts of the cannabis during the intervals -ñ

In the years that followed, Cannabis came to be perceived as a drug of abuse, smoked by certain classes of people as "marijuana" or "marihuana." In an article entitled "The Weed of Insanity" (Bragman, 1925), the author admitted, "It has some value in the relief of migraine."
The following year, another author of a popular textbook remained a convinced user of Cannabis for migraine (Stevens, 1926):

Cannabis indica is sometimes very useful, when a reliable preparation can be secured. -Morphin should never be employed, except as a last resort.

Despite its political downturn in popularity, a 1933 review of therapeutic techniques (Fantus, 1933) recommended "fluidextract of cannabis":
One teaspoonful in water every two hours until relieved. (For migraine.)

In 1937, marijuana was rendered essentially illegal in the U.S.A. after a contrived set of congressional hearings. Cannabis had become a phytochemical scapegoat for a perceived social problem, and research on its medical uses was substantially curtailed. The American Medical Association vigorously opposed this development (Cary, 1937):

There is positively no evidence to indicate the abuse of cannabis as a medicinal agent or to show that its medicinal use is leading to the development of cannabis addiction. Cannabis at the present time is slightly used for medicinal purposes, but it would seem worth while to maintain its status as a medicinal agent for such purposes as it now has. There is a possibility that a restudy of the drug by modern means may show other advantages to be derived from its medicinal use.

Despite this political event, in 1938 Robert Walton published a comprehensive review of Cannabis, with botanical, historical, chemical and political discussions (Walton, 1938). After discussing the issues of its abuse, and consequent legislation, he stated:

More stringent regulations making the drug unavailable for medical and scientific purposes would be unwise, since other uses may be developed for the drug which will completely overshadow its disadvantages. The drug has certain remarkable properties and if its chemical structure were determined and synthetic variations developed, some of these might prove to be valuable, both as therapeutic agents and as experimental tools.

In 1941, Cannabis preparations were dropped from the United States Pharmacopoeia (U.S.P.), but the following year, the editor of the Journal of the American Medical Association still advocated oral preparations of Cannabis in treatment of menstrual (catamenial) migraine (Fishbein, 1942):

In this instance the patient may be given either sodium bromide or fluidextract of cannabis three days before the onset of the menstrual period, continued until three days after the menstrual period.

As a seeming afterthought, he added:

Ergotamine tartrate may also be given.

The latter medicine remains in the migraine armamentarium, fifty-six years later, but he considered it inferior to Cannabis.
Thus, as we have seen, Cannabis was touted in eight consecutive decades in the mainstream Western medical literature as a, or the, primary treatment for migraine.
In 1944, a perception of rising crime and addiction due to Cannabis was feared in urban America. The mayor of New York authorized an examination of the issue by the New York Academy of Medicine (La Guardia Commission, 1944). Among its conclusions were the following:

The practice of smoking marihuana does not lead to addiction in the medical sense of the word.
The use of marihuana does not lead to morphine or heroin or cocaine addiction and no effort is made to create a market for these narcotics by stimulating the practice of marihuana smoking.
Marihuana is not the determining factor in the commission of major crimes.
Marihuana smoking is not widespread among school children.
Juvenile delinquency is not associated with the practice of smoking marihuana.
The publicity concerning the catastrophic effects of marihuana smoking in New York City is unfounded.

Despite political issues in the U.S.A., medical use of Cannabis continued elsewhere. In 1947, an ethereal extract of Cannabis was still employed for migraine treatment in Argentina (Kabel"k, Kreje" & Santavy, 1960). Cannabis was still recommended as a homeopathic remedy for migraine in 1956 in then East Germany (Auster & Schafer, 1956).
In Tashkent in the 1930's, Cannabis or nasha was still employed medicinally including an indication for headache, despite Soviet prohibition (Benet, 1975).
Similarly, Cannabis remained a useful item in the pharmacopoeia of China (Stuart, 1928). Perry (1980) referred to ongoing use of Cannabis in China to treat migraine, much the same as noted in Thailand (Ponglux et al., 1987). In other areas of Southeast Asia its use remains popular (Martin, 1975):

Everywhere it is considered to be of analgesic value, comparable to the opium derivatives. Moreover, it can be added to any relaxant to reinforce its action. Cooked leaves, which have been dried in the sun, are used in quantities of several grams per bowl of water. This decoction helps especially to combat migraines and stiffness; taken before sleep and before meals, it relaxes the nerves.

Analgesic effects of Cannabis have retained importance in the folk medicine of North Africa (Boulos, 1983).
As late as 1957, despite governmental regulation in India, Cannabis drugs retained a role in its indigenous medicine (Chopra & Chopra, 1957):

The concentrated resin exudate - is considered valuable in preventing and curing sick-headaches, neuralgias and migraine

The following year, in a treatise entitled Indigenous Drugs of India (Chopra et al., 1958) the authors stated:

Cannabis is used in medicine to relieve pain, to encourage sleep, and to soothe restlessness. There is little definite knowledge of the therapeutic effects produced, but in some persons it appears to produce euphoria and will often relieve migraine headaches.
-The safest way of determining the dose of an individual preparation is to give it in ascending quantities until some effect is produced.

In discussing the native use of Cannabis and opium products by village doctors in India, who provided 80% of the population with their medical care, the author of a report to the United Nations felt a legitimate role for them was still present (Dwarakanath, 1965):

These drugs should be allowed to be used by Ayurvedic and Unani [Arabian tradition] physicians until such time as the benefits of modern medicine are extended to rural areas. Banning their use by the large mass of Ayurvedic and Unani physicians for therapeutic purposes may create a vacuum which may not be easily filled for a long time to come.

In another, undated book about medicinal plants of India (Dastur, n.d.), the author stated:

Charas is the resinous exudation that collects on the leaves and flowering tops of plants it is the active principle of hemp; it is a valuable narcotic, especially in cases where opium cannot be administered; it is of great value in malarial and periodical headaches, migraine -

In recent decades, marijuana has moved to center stage of Western consciousness, not as a medicinal agent, but rather as a perceived drug of abuse and gateway to more harmful addictive substances. Research resumed only slowly, with occasional anecdotal reports by patients of Cannabis' benefits on their illnesses.

B.2 More Recent Research and Political Developments on Cannabis:

The respected journal Science published an article entitled "Clinical and Psychological Effects of Marihuana in Man" in 1968. It examined a variety of clinical responses of nine volunteers smoking Cannabis under clinical conditions. Some marijuana-naive subjects noted little subjective change, but manifested some degree of impairment on simple cognitive tests. Some regular users of the drug actually improved their performance over that in the sober state. Moderate heart rate increases were noted, as was conjunctival redness. Peak drug effects were noted within thirty minutes, and were gone in three hours (Weil, Zinberg & Nelsen, 1968).
In 1974 began a series of studies that examined analgesic effects of Cannabis. The first article (Noyes & Baram, 1974) described case studies of five patients who voluntarily employed it to treat their painful conditions. Three of these had chronic headaches, with definite, but mixed responses. Of "Case 2" it was stated:

A 24-year old single candidate for a master's degree in creative writing claimed to have used marijuana for migraine headaches for a year and a half. At the unofficial suggestion of a neurologist, he experimented with the drug and, finding it effective, used it in lieu of ergotamine tartrate on six or more occasions. Each time he claims to have prevented or substantially attenuated a headache. Those that developed lasted less than 4 hr and were mild in intensity. He had been particularly aware of the relaxing effect of the drug because migraine attacks generally aroused considerable anxiety within him. The effects of smoking for migraine were different from those experienced with a comparable dose on social occasions. Euphoria was absent and drowsiness developed, which helped him to sleep and thus avoid further symptoms. He rated Cafergot PB (with phenobarbital) as slightly superior to marijuana in preventing headaches [This preparation has been particularly prone to provoke "analgesic rebound" with promotion of headache recurrence]. During the past year he claimed to have diminished the frequency of migraine attacks by smoking.

The authors said of "Case 3":

A 30-year-old married housewife claimed to have used marijuana for the relief of headaches and other minor discomforts for over a year. She discovered its value while smoking and, coincidently, suffering from a headache. She described immediate and lasting relief from a dose similar to that used on social occasions. This was associated with a reduction in tension. In her experience, marijuana had been superior to aspirin, the only other medication she had used.

Of "Case 5" they stated:

A 25-year-old single graduate student discovered, on the recommendation of a friend, that marijuana relieved headaches. She reported smoking the drug occasionally for that purpose for more than 2 years. Though often complete, the relief she obtained was not consistent. She believed that she had benefited from smoking only about 70% of the time [comparable to the best standard pharmaceuticals in 1997].

Subsequent experimental studies explored these reported analgesic effects of Cannabis. One article examined pain tolerance thresholds (Milstein et al., 1975). Both native (8% increase) and experienced subjects (16% increase) noted statistically significant increases in pain threshold after smoking Cannabis.
Another study pertained to oral THC in cancer patients (Noyes et al., 1975a). Pain relief with escalating doses significant to the P<0.001 level was observed. Peak effects occurred at three hours with doses of 10 and 15 mg., but were delayed until five hours after the 20 mg. oral dose.
A similar research group compared the analgesic effect of THC was compared to codeine (Noyes et al. 1975b). In short, 10 mg. of oral THC reduced subjective pain burdens by similar decrements to 60 mg. of codeine, as did 20 mg. of THC vs. 120 mg. of codeine. Subjects tolerated 10 mg. of THC well tolerated, but 20 mg. produced sedation and psychic disturbances in some. It should be stated that patients were relatively elderly, and not previously accustomed to the effects of the drug.
In the 1980's research in the pharmacology of Cannabis and its derivative THC continued. In 1983, one study examined varying potencies of smoked Cannabis and showed correlation between serum THC levels and subjective "high" (Chiang & Barnett, 1983). Experimental subjects accurately rated potency of the various samples. In another article (Wall, Sadler & Brine, 1983), no differences were identified in metabolism and kinetics of THC in men and women.
In a forensic review (Mason et al., 1985), marijuana's effect on driving was addressed:

Sparse as the reports may be, they tend to show that if there are drivers who are unsafe because of marijuana, their numbers are small and most are also influenced by alcohol.

The authors also noted no correlation between plasma THC levels and observed driving impairment.
In "Cellular Effects of Cannabinoids" (Martin, 1986), the author reported that naloxone did not block the analgesic properties of these substances, supporting a non-opioid mechanism.
In an article on pharmacokinetics (Agurell et al., 1986), findings included the observation that smoking Cannabis destroyed 30% of available THC. Additionally, it was reiterated that standard laboratory chromatographic techniques lacked sufficient sensitivity to measure serum THC levels in man.
The final article, "Health Aspects of Cannabis" (Hollister, 1986), made a several pertinent points:

The efficiency of the delivery of a dose by smoking has been estimated to be about 18%, but frequent smokers obtain 23% while infrequent users obtain only 10% (Lindgren et al., 1980). THC and marijuana extracts are also active by mouth; the systemic bioavailability of oral administration is only about 6%, one-third that from smoking (Ohlsson et al., 1980). -Oral doses delay the onset of symptoms for 30 min to over 2 h, as well as prolonging the span of action of the drug.

In reviewing a study involving smoking of massive doses daily for a prolonged period, findings included lower intraocular pressure, serum testosterone levels, and airway narrowing, but no chromosomal aberrations, or impairment of immune responses were noted (Cohen, 1976).
Various "marijuana myths" were debunked on the basis of literature review. Much as reported by prior commissions, while aggravation of pre-existing psychotic conditions by marijuana use was observed, no etiologic role of Cannabis was supported. Chronic use in Jamaica (Comitas, 1976) and Costa Rica (Carter & Doughty, 1976) revealed normal worker production and motivation in what would be considered blue-collar jobs. Two computerized tomography (CT scan) studies failed to support previous reports of cerebral atrophy after heavy use (Co et al., 1977; Kuehnle et al., 1977).
On issues of behavior, Hollister (1986) stated:

The myth dies hard that marijuana makes otherwise docile subjects violent. Virtually every experimental study of cannabis that has tried to measure violent or aggressive behavior or thoughts during cannabis intoxication has come to the same conclusion; they are decreased rather than increased.

Of addiction, he wrote:

In man, a somewhat similar, though mild, withdrawal reaction was uncovered after abrupt cessation of doses of 30 mg of THC given every 4 h p.o. for 10 to 20 days. Subjects became irritable, had sleep disturbances, and had decreased appetite. Nausea, vomiting and occasionally diarrhea were encountered. Sweating, salivation and tremors were autonomic signs of abstinence (Fink et al., 1976). Relatively few reports of spontaneous withdrawal reactions from suddenly stopping cannabis use have appeared despite the extraordinary amount of drug consumed. Five young persons experienced restlessness, abdominal cramps, nausea, sweating, increased pulse rate, and muscle aches when their supplies of cannabis were cut off. Symptoms persisted for 1 to 3 days (Benusan, 1971). The rarity of reports of these reactions may reflect the fact that they are mild-

Hollister (1986) ultimately addressed possible medical indications, but his experience with migraine was not broad:

Migraine: This indication has not been studied systematically in recent years, although it has a long history. In one patient I treated, the mental effects sought socially caused the patient to abandon treatment.

The following year, another article dealt with the issue more directly (El-Mallakh, 1987). Entitled "Marijuana and Migraine," three cases were discussed in which abrupt cessation of frequent, prolonged, daily marijuana smoking were followed by recurrent migraine attacks. One patient noted subsequent remission of headaches with a return to episodic marijuana use, while the two others employed "conventional drugs" successfully. THC's peripheral vasoconstrictive actions in rats, or its action to minimize serotonin release from the platelets of human migraineurs (Volfe, Dvilansky & Nathan, 1985), were felt to be possible explanations of its therapeutic effects.
In 1988 marijuana's status as a Schedule 1 drug with no therapeutic value was challenged. A reclassification to Schedule 2, might make marijuana potentially available for prescription to patients. Francis Young, the DEA administrative law judge, reviewed the voluminous data from patients, scientists, and politicians. He approved its use as an anti-emetic, and anti-spasticity drug in multiple sclerosis and found its utilization in glaucoma reasonable. Pertinent excerpts follow (Young, 1988):

By any measure of rational analysis marijuana can be safely used within a supervised routine of medical care.

-There have been occasional instances of panic reaction in patients who have smoked marijuana. These have occurred in marijuana-naive persons, usually older persons, who are extremely anxious over the forthcoming chemotherapy and troubled over the illegality of their having obtained the marijuana. Such persons have responded to simple person-to-person communication with a doctor and have sustained no long term mental or physical damage.

Marijuana should not be used by persons anxious or depressed or psychotic or with certain other health problems.

Cannabis has also been reviewed in the French literature (Spadone, 1991). Among other points, the author indicated:

As to serotonin, the synthesis of 5-HT is stimulated by THC (possibly by intermediary augmentation of corticosteroids) as well as brain 5-HT content. Synaptosomal uptake seems inhibited, while release is favored. [translation EBR]

In 1992, subjective preferences of experimental subjects smoking Cannabis, or ingesting oral THC were assessed (Chait & Zacny, 1992):

All ten subjects on both trials chose active marijuana over placebo, a proportion significantly greater than expected by chance (P<0.01).

Oral THC was chosen [over oral placebo] on both trials by 10 of the 11 subjects (P<0.02).

These results serious call into question the plausibility of true blinding with placebo preparations in prospective therapeutic drug studies, especially those with smoked marijuana.
In recent years, scientists have provided elucidation of the mechanisms of action of Cannabis and THC with the discovery of an endogenous cannabinoid brain receptor, arachidonylethanolamide, or anandamide (Barinaga, 1992; Devane et al., 1992; Marx, 1990; Matsuda et al., 1990). Anandamide has an inhibitory effect on cyclic AMP in target cells, which, though widespread in the brain, cluster in nociceptive areas (Herkenham, 1993). Preliminary tests of its action behavioral activity support similarity to THC (Fride & Mechoulam, 1993).
Subsequently, additional research has pondered mechanisms of therapeutic action of the cannabinoids on migraine. Anandamide and other cannabinoid agonists inhibit rat serotonin type 3 (5-HT3) receptors (Fan, 1995). This receptor acts as a mediator of emetic and pain responses. In 1996, researchers demonstrated antinociceptive effects of delta-9-THC and other cannabinoids in the periaqueductal gray matter in rats (Lichtman, Cook & Martin, 1996). The PAG is a putative migraine generator area (Goadsby & Gundlach, 1991; Raskin, 1988).
Another study employing the cannabinoid agonist WIN 55,212-2 has demonstrated a specific antinociceptive effect in the ventral posterolateral nucleus of the thalamus (Martin, Hohmann & Walker, 1996). This demonstrates an additional possible mechanism of cannabinoid-induced analgesia.
While these developments indicate some of the mechanisms of Cannabis and THC effects on natural cerebral biochemical processes, political debate has intensified. In 1993, a book entitled Marihuana: The Forbidden Medicine (Grinspoon & Bakalar, 1993) included an entire section on migraine. One clinical vignette documented the medical odyssey of a migraineur through failures with standard pharmaceuticals:

Several years later the migraines returned, and my husband said he had read that marihuana was good for headaches. I was amazed. Two hits and a short rest completely warded off the nausea and headache. As soon as I noticed the flickering visuals that forewarned me of an approaching migraine, I could take a little cannabis and a short nap and the migraine would not develop at all. I was usually ready to go back to work in half an hour. It gave me a feeling of tremendous power to be finally in such control of my migraines.
In the eighteen years since I began using cannabis to relieve migraines, I have been caught away from home several times without my herb. Once I tried taking Tylenol and found it helped a little with the pain but not at all with the nausea or the visual effects. Both of my daughters (now seventeen and twenty-one) also get occasional migraines, which first appeared when they began to menstruate. Both get tremendous relief from cannabis herb. My mother suffers from severe headaches, but she has never used cannabis because it is illegal.

The Journal of the American Medical Association published a recent impassioned commentary (Grinspoon, 1995):

Marihuana is also far less addictive and far less subject to abuse than many drugs now used as muscle relaxants, hypnotics, and analgesics. The chief legitimate concern is the effect of smoking on the lungs. Cannabis smoke carries even more tars and other particulate matter than tobacco smoke. But the amount smoked is much less, especially in medical use

The main active substance in cannabis, [delta-9]-tetrahydrocannabinol ([delta-9]-THC) has been available for limited purposes as a Schedule II synthetic drug since 1985. This medicine, dronabinol (Marinol), taken orally in capsule form, is sometimes said to obviate the need for medical marihuana. Patients and physicians who have tried both disagree. The dosage and duration of action of marihuana are easier to control, and other cannabinoids in the marihuana plant may modify the action of [delta-9]-THC. The development of cannabinoids in pure form should certainly be encouraged, but the time and resources required are great and at present unavailable. In these circumstances, further isolation, testing, and development of individual cannabinoids should not be considered a substitute for meeting the immediate needs of suffering people.

The American Journal of Public Health issued a particularly strong plea (AJPH, 1996), quoted in excerpts:

9513: Access to Therapeutic Marijuana/Cannabis

Understanding that while synthetic tetrahydrocannibinol (THC) is available in pill form, it is only one of approximately 60 cannabinoids which may have medicinal value individually or in some combination; and
Understanding that marijuana has an extremely wide acute margin of safety for use under medical supervision and cannot cause lethal reactions; and
Understanding that marijuana has been reported to be effective in: - e) decreasing the suffering from chronic pain; -and
Understanding that marijuana seems to work differently than many conventional medications for the above problems, making it a possible option for persons resistant to the conventional medications; -
Concluding that greater harm is caused by the legal consequences of its prohibition than possible risks of medicinal use; therefore
1. Encourages research of the therapeutic properties of various cannabinoids and combinations of cannabinoids; and
2. Encourages research on alternative methods of administration to decrease the harmful effects related to smoking; and
3. Urges the Administration and Congress to move expeditiously to make cannabis available as a legal medicine where shown to be safe and effective and to immediately allow access to therapeutic cannabis through the Investigational New Drug Program.

In a current review of over 65,000 patient records in an HMO (Sidney, Beck & Tekawa, 1997), it was said:

In summary, this study showed little, if any, effect of marijuana use on non-AIDS mortality in men and on total mortality in women.

Marijuana myths have recently been exhaustively reviewed in book form (Zimmer and Morgan, 1997).
In a recent review article (Voth & Schwartz, 1997), another viewpoint on Cannabis safety and efficacy was presented:

The evidence does not support the reclassification of crude marijuana as a prescribable medicine.

However, their study confined itself to the clinical issues of nausea, appetite stimulation, glaucoma, and spasticity. Methodologically, it is open to criticism in that the authors screened only the 1975-1996 medical literature, an era during which clinical research on therapeutic benefits of Cannabis was next to impossible. The authors did not examine migraine as an issue, failed to review the extensive literature of the past, and ignored editorial comments and anecdotal accounts by patients.
Recently, the debate on the subject of "medical marijuana" has extended to the World Wide Web. One posted document (Mikuriya, 1996) is "Chronic Migraine Headache: five cases successfully treated with Marinol and/or illicit cannabis." Two patients were prescribed dronabinol for their headaches with improvement, but some degree of side effects, or difficulties with overwhelming cost. Both switched to marijuana, with an improved clinical response and decreasing frequency and severity of migraine attacks. Another family of three women smoked marijuana acutely with good success in aborting headache, often in the prodromal phase. Another Web document entitled "Cannabis Medicinal Uses at a 'Buyers' Club'" examined the indications that prompted patients to seek out this treatment. Of the 57 people interviewed, 11 identified migraine as the culprit condition that prompted their decision to self-medicate with Cannabis.
Alternative smoke delivery systems have been investigated for Cannabis (Gieringer, 1996a and b). Reportedly, vaporization of marijuana makes it possible to deliver even high doses of THC to the lungs of a prospective patient far below the flash point of the Cannabis leaf, thus reducing smoke, tar and other possible carcinogens. However, the standard marijuana joint remained about as effective as any examined smoking device, including those employing water filtration, in providing a favorable ratio of THC to tar and other undesirable by-products. A standardized smoking procedure for use of Cannabis in medical research has been described (Foltin, Fischman & Byrne, 1988).
As we have seen, suppository preparations of Cannabis have been used to advantage in the past, and may be an acceptable alternative route of administration for the migraineur, although the advantage of dose titration would be lost.


B.3 Conclusions:

Cannabis, taken orally or inhaled, has a long history of reportedly safe and effective use in the treatment and prophylaxis of migraine.
Cannabis has demonstrable analgesic effects.
Cannabis affects nociceptive pathways in the brain, and may interact with serotonergic mechanisms deemed important in the migraine cascade.
Cannabis is an effective anti-emetic, which is a useful property in migraine treatment.
Cannabis, even when abused, has minimal addiction potential, and seems to be safe for occasional usage, particularly if monitored by a physician.
Cannabis' primary problem as a medicine lies in its possible pulmonary effects, which seem to be minimal in occasional, intermittent use. This danger could be reduced further with the selective breeding of Cannabis with a higher THC content.
Cannabis when smoked, is rapidly active, obviates the need for gastrointestinal absorption (impaired markedly in migraine), and may be titrated to the patient's needs for symptomatic relief, and to his or her tolerance.
Cannabis, when smoked in the form a marijuana cigarette, or "joint," presents the hypothetical potential for quick, effective parenteral treatment of acute migraine.

 

C. Preliminary Studies/Progress Report

The principal investigator, Ethan Russo, M.D., has practiced clinical neurology for fifteen years. He has had a long interest in the study and treatment of migraine, and is the Medical Director of the St. Patrick Hospital Inpatient Pain Treatment Program.
In 1990, he continued a personal investigation of herbal medicine by embarking a world survey of available ethnobotanical information with reference to headache treatment. Also in that year, publications outlining the biochemical basis of migraine treatment in serotonin receptor pharmacology suggested to him the plausibility of employing these laboratory techniques as an in vitro method of screening plant extracts for possible development as novel pharmaceuticals (Humphrey et al., 1990; Peroutka, 1990). In 1992, he published an article in Journal of Ethnopharmacology (Russo, 1992)(Appendix), outlining these theories, and how they might be applied to 24 plants used for headache treatment by indigenous peoples of the Ecuadorian Amazon. A field study of these plants with collection and subsequent laboratory analysis was proposed, and deemed worthy of inclusion in Spirit of Enterprise: The 1993 Rolex Awards (Appendix).
Subsequently, two University of Montana Research Grants, totaling $5000 in awards, were assigned to investigations of plant candidates from the countries of Peru and Belize.
In 1995 the PI accomplished two months of ethnobotanical field study of the Machiguenga tribe in the remote Parque Nacional del Man? of Peru.
On expedition, he and his research partner collected in excess of 400 medicinal plant species for botanical identification. Of these, fully 25% had indications suggesting psychopharmacological activity, including migraine. About 90 species were preserved in ethanol, and in some instances, live specimens were exported in accordance with Peruvian and U.S. statutes. Preliminary investigation confirms that very few have undergone any biochemical analysis. Further details of this work are available at the Internet Web Site:
"Plants of the Machiguenga": http://www.montana.com/manu
Initial studies of these specimens to date, undertaken by colleagues at the University of Montana, have revealed very encouraging results in several plant extracts on serotonin receptor activity (Russo et al., 1997) (Appendix). These hold obvious promise for additional study in migraine treatment. This team has continued investigation of other plants of reported migraine benefit including feverfew, Tanacetum parthenium (Weber et al., 1997)(Appendix).
The American Gloxinia and Gesneriad Society, and the Multidisciplinary Association for Psychedelic Studies have provided small grants to Dr. Russo for investigation of the gesneriad family, and plants with ethnobotanical evidence for migraine treatment efficacy, respectively.
Dr. Russo holds a current Schedule 1 Drug Permit (Appendix), and is thus licensed to use each of the study medications, should this project be approved. A proposal by Dr. Russo to examine the activity Cannabis and dronabinol on serotonin receptor activity has been approved by DEA and NIDA (Russo et al., 1997), and is currently underway.
With the PI's extensive clinical experience in migraine and treatment of medication side-effects, and the expertise of the study team in data and statistical analysis, we are confident of achieving the stated goals of this project to investigate the putative role of dronabinol and smoked Cannabis in acute headache treatment.

D. Research Design and Methods

D.1 Procedure and Subject Activities:
Potential subjects will be recruited via advertisements (enclosed-Appendix), or via referral by local emergency departments, or private physician offices. Prospective patients will contact one of the researchers for an initial interview by telephone. The researcher will follow the attached protocol (Appendix) for interviewing prospective subjects, and use established exclusion criteria below noted. At the end of the interview, potential subjects that fit International Headache Society (IHS) criteria for migraine with or without aura (Headache, 1988) and do not merit exclusion will be informed that Cannabis (marijuana) would be one of the treatments used within the study. Selected patients will hopefully include and mixture of Cannabis-experienced and Cannabis-naÔve individuals. We will explain how subjects will be treated for acute migraine with study drugs in each of up to ten sessions in each of two three-month blocks with a 2-week washout period in between. The procedure for administration of study drugs will be explained in detail. All patients will undergo a detailed history and screening neurological examination by Dr. Russo to exclude space-occupying lesions or other causes of headache.
Before the first session, the subjects would be asked to complete a consent form (approved by the St. Patrick/Community Hospital Joint IRB)(Appendix). Following agreement to participate in this study, patients will call in to arrange treatment of an acute migraine. Each subject will be randomized to Group A or Group B by coin toss. The study will employ a "double dummy technique," as suggested by the Ad Hoc Group of Experts of the Workshop on the Medical Utility of Marijuana convened by the NIH in 1997 (Appendix). One group will begin the first three-month block receiving either dronabinol 10 mg. p.o. plus smoke a placebo marijuana cigarette, or (by coin toss) receive sumatriptan 6 mg. s.c. The other group in the first 3 month block will receive either sumatriptan 6 mg. s.c., or (by coin toss) a placebo capsule resembling dronabinol plus smoke a 4% THC-content marijuana cigarette. Patients who have severe discomfort with injected sumatriptan, but lack true allergic sensitivity may alternatively receive nasal sumatriptan administration (20 mg.). In each smoking session, patients will be administered one or more inhalations, titrated to their seeming clinical response and comfort, to a limit of one whole cigarette employing a modified "uniform puff procedure"(Foltin et al., 1988):
a) Study subjects will inhale the marijuana cigarette for five seconds.
b) They will hold the smoke in for ten seconds before exhaling.
c) They will wait thirty seconds before resuming the cycle.

After smoking, the number of inhalations taken and remaining butt length and weight of the marijuana cigarette will be measured and recorded. Dr. Russo or the study nurses will supervise administration of all medications.
Following the study drug treatment, each subject will be asked to remain in the study room to rest. The nursing staff will also monitor behavior and assure that the subject has not had adverse reaction to the medication. After 60 minutes, the subject would meet with study personnel to undergo a Folstein Mini-Mental State Examination (Folstein et al., 1975)(form-Appendix), and complete a questionnaire employing visual analogue scales regarding their symptoms and level of function (Appendix). Blood samples will be drawn for quantitative determination of THC levels in the dronabinol and marijuana sessions at 10 minutes and two hours, in accordance with previous data on pharmacokinetics of THC discussed in Section B. Any patient who remains uncomfortably symptomatic with migraine after three hours will have the option of receiving magnesium sulfate 1000 mg. IV (Mauskop et al., 1996). This was chosen due to its combination of efficacy without drug interaction concerns.
The Mini-Mental State and questionnaires will be repeated at the two, three and four-hour marks post-treatment. In each marijuana and dronabinol session, patients will be asked to judge what medicine they received.
The subjects will then be given a stamped, self-addressed envelope containing additional questionnaires for completion (Appendix) at 24 hours to mail back to the researchers.
Before the next treatment sessions are administered, subjects will be asked the questions on the Pretreatment Questionnaire (Appendix). The same protocol described above would be administered for each of the study drug sessions, to a maximum of ten sessions per three-month block.
After three months, a two-week "washout period" will be observed, and then the two groups will switch. Thus those who previously received active Cannabis/placebo dronabinol or sumatriptan will subsequently receive active dronabinol/placebo Cannabis or sumatriptan and vice versa.
Through the use of overlapping schedules, study sessions will be performed from 07:00-21:00, Monday through Saturdays, thus optimizing patient availability and data acquisition. This regimen should be adequately reasonable, given that most migraines are generated in A.M. hours. Patients will be encouraged to call early in their attacks to arrange treatment. We do not feel that it is reasonable to hospitalize patients, inasmuch as this will markedly elevate study costs, but more importantly, inhibit recruitment.


D.2 Statistical Design, Analysis and Sample Size Calculations
by Brian M. Steele, Ph.D.

The statistical design of the study is crossover design. The strength of the crossover design is that subjectspecific variation may be estimated and accounted for, and the design permits manifestation of cumulative drug effects. Under the crossover design, each subject is placed on a single protocol for a fixed time period (a session), during which a number of visits for migraine treatment occur, and then the subject is switched to the alternate protocol. The proposed design specifies that 20 subjects will be randomly allocated to each protocol and that 7 visits per protocol are expected, based on the average frequency of migraine attacks for patients in the study population. Visits per 3-month protocol are limited to 10.
The study population is adult acute migraine patients meeting IHS criteria. The two protocols are described above. The primary response variable is a selfrated measure of pain. The scale is 110 points, and the measurement is made hourly after each treatment. Additional response variables will also be measured hourly after treatment, and numerous covariates will be measured.
The problem of subject dropout and variable numbers of observations from each subject are resolved by using linear mixed model and associated likelihoodbased inferential methods. The linear mixed model approach avoids the necessity of a balanced experimental design required for traditional analysis of cross-over designs using analysis of variance methods. Hence, while there is a potential loss of power from dropout, uneven numbers of observations from subjects does not substantial degrade the power of the design.
The study focus is on characterizing patient response to pyrolysed Cannabis. The primary variables of interest are selfrated pain, nausea, and sensitivities to light and sound hourly after administration of study medications. A secondary response variable is the time elapsed between migraine attacks. Response variables will be analyzed individually, with care taken to avoid multiple testing problems and to control experimentwise error rates. The linear mixed model approach is taken, as it is appropriate for cross-over designs in which there are variable numbers of observations from each subject. Furthermore, the linear mixed model easily allows for estimation and control of covariates in the analysis. The principal objectives of the statistical analysis are estimation and comparison of treatment effects, and secondarily, assessment of covariate effects. Aside from descriptive statistics, the linear mixed model approach will be used to accomplish these objectives.
Sample size calculations are based on a test of the null hypothesis stating that there are no differences between the mean responses for the oral dronabinol and cannabis treatment groups, versus the alternative hypothesis stating that the mean responses differ by 1.5 point on the 0 to 10 point pain awareness scale. It is also assumed that within and between subject standard deviations are equal and 3 on the 0 to 10 scale, and that the treatment means were 2.5, 6.5, and 5 for the sumatriptan, oral dronabinol, and Cannabis groups, respectively. These assumptions should be considered as educated, but conservative, guesses aimed at insuring adequate sample sizes. Given the assumptions, simulated data were generated for various combinations of numbers of subjects and visits, and twenty percent of the observations for each data set were deleted at random to simulate missing data and dropouts. One hundred fifty simulated data sets were generated for each combination of subjects and visits and, for each, the test for differences between the oral dronabinol and cannabis treatment groups was carried out. The fraction of tests that were significant at the 95% level is the estimated power for that combination of subjects and visits. Figure 1 (page 47) shows approximate contours of equal power for a range of numbers of subjects and visits, or trials. It reveals that 20 subjects per protocol and 6 visits per protocol will yield a approximate power of 0.99. In this scenario, there are 40 subjects in the study (randomly allocated to one of two protocols) and 6 visits per subject per protocol are planned on. The sample size calculations are based on 20% fewer visits (i.e., an average of 4.8 visits per protocol) as 20% of the simulated data were deleted at random. In conclusion, 20 subjects per protocol and 7 planned visits per protocol will insure that a reduction in pain severity of 1.5 on the 10 point scale will be detected at the 95% confidence level.

D.3 Location of Research:

The administration of these medications will take place in the Neurosciences Department at the Western Montana Clinic in space rented from St. Patrick Hospital. The Joint IRB has accepted this plan (Appendix). Precautions described below will be employed to manage risks to patients, and reduce ambient smoke exposure. Hours of availability are discussed above, Section D.1.

D.4 Benefits:

Results of this project may be valuable for migraineurs and the professionals who treat them because there is a strong need for additional medications that will effectively this condition in its acute state. At this time, the best available medication, injected sumatriptan (Imitrex) has been ineffective in up to 30% of patients, or has produced undesirable side effects for up to 66% when administered subcutaneously (Mathew, 1997). Sumatriptan is the current "gold standard". Sumatriptan non-responders have been studied (Visser et al., 1996). Such patients may be obese, or take the medicine too early. Beyond that, the study found no features distinguishing responders from non-responders. The PI would add another observation from clinical experience: people with the chronic daily headache subset of migraine respond poorly to subcutaneous sumatriptan. Because this proposal focuses on migraineurs with episodic attacks, CDH patients will be excluded.
Despite the development of serotonin 1D-agonist medications, migraine remains a serious public health issue. An estimated 23 million Americans suffer severe migraine. Of these, 25% have four or more episodes per month, and 35% have one to three severe headaches each month (Stewart et al., 1992). In economic terms, the impact of migraine is enormous: an estimated 14% of females, and 8% of males missed a portion of, or an entire day of work or school in one month (Linet et al., 1989). Migraine has been estimated to account for an economic impact of $1.2 to $17.2 billion annually in the U.S.A. in terms of lost productivity (Lipton et al., 1993). For this reason, new treatment alternatives need to be explored and evaluated for efficacy.
The principal investigator also believes that the issue of medical marijuana deserves proper scientific examination. In modern times, controlled studies of Cannabis for clinical treatment of diseases have not been permitted. Given the extensive "anecdotal" documentation of past efficacy of Cannabis by the premier physicians of the times, and the intense scrutiny of studies indicating its safety, modern studies employing controlled techniques are warranted.

D.5 Gender and Minority Inclusion:

This study will be open to all adults subject to the medical exclusions noted above. We encourage participation of women who are not pregnant or lactating, especially considering that women suffer a higher incidence of migraine in most studies. Efforts will be made to ensure reasonable birth control methods in participating women in child-bearing years. The local population of Missoula, MT contains small minorities of Native Americans and Hmong refugees. All will be welcome to participate.

E. Human Subjects

E.1 Population Characteristics:
Please see section D.1 above. It is anticipated that the study population will consist of healthy, mostly young adults with migraine. Forty subjects with an interest in completing the full 6 1/2-month course of up to 20 study sessions will be chosen. Age range will be 18 and upward. As discussed below, women and minorities wil l be encouraged to take part in the study, and subjects will include experienced and naÔve subjects with respect to Cannabis.
Migraine afflicts 14% of females, and 8% of males (Linet et al., 1989) for a composite of 11%. Of those, 25% are severely affected (Stewart et al., 1992), or 2.75%. Multiplying that by an estimated adult population in the Missoula study area, we have: 60,000 x 0.0275 = 1650 potential subjects available.

E.2 Research Material and Data:
Serum THC samples will be collected from study patients, and new data will be generated in the form of questionnaires for statistical analysis.

E.3A Recruitment: please see D.1 above.

E.3B Informed Consent:
During the intake interview, study subjects will receive full information about the prospective use of Cannabis, and the other study drugs, with the option to decline participation. Upon agreeing to enroll in the study at a rate of reimbursement of $30 per study session, they will be asked to sign a consent form (approved by the Joint IRB)(Appendix). Because of the sensitive political nature of this investigation, it is our preference that suitable discretion be employed by all participants, and not discussed with other than appropriate individuals prior to its completion.

E.4 Potential Risks:
The Australian government (Hall et al., 1995) compiled a recent exhaustive review of sequelae of Cannabis use. In the summary, it states:

Acute Effects
-- anxiety, dysphoria, panic and paranoia, especially in native users;
-- cognitive impairment, especially of attention and memory, for the duration of intoxication;
-- psychomotor impairment, and probably an increased risk of accident if an intoxicated person attempts to drive a motor vehicle, or operate machinery;
-- an increased risk of experiencing psychotic symptoms among those who are vulnerable because of personal or family history of psychosis;
-- an increased risk of low birth weight babies if cannabis is used during pregnancy.

For these reasons, pregnant or lactating women, those with past or family history of psychosis, patients with coronary or hypertensive risk factors, patients taking MAO-inhibitors or having previous problems with substance or analgesic abuse, etc., will be excluded form participation in the study. Patients with the chronic daily headache variant of migraine will also be excluded, as will those with history sumatriptan allergy or significant chest pain. Patients will also undergo a detailed history and physical examination by Dr. Russo to screen for additional exclusions. As above, efforts will be made to ensure reasonable birth control methods in study subjects of child-bearing potential.
There is the possibility that marijuana-naÔve subjects will experience some pulmonary discomfort. This will be minimized by instruction in techniques of inhalation discussed previously. Anxiety reactions, if they occur, will be managed through personal reassurance, and soothing music.
For these reasons, all subjects will remain for four hours after initial treatment. The subjects will be asked about the availability of a designated driver for their sessions. No subjects will be allowed to drive themselves following treatment. If no designated driver is available, other transportation will be provided via taxi, or study personnel. Dr. Russo or a delegate will be available "on call" subsequently after study-patients return home. Each patient will have contact numbers available throughout the study period to reach him or a delegate.
Study patients will be instructed that may employ oral ibuprofen or naproxen as "rescue medication" if needed, but use of other medicines with analgesic rebound or psychoactive potential will be discouraged. Concomitant use of migraine prophylactic drugs will not necessarily be an exclusion in this study, so as to more closely mirror potential "real world" issues. It is anticipated that most study subjects will not be taking such medication. Recreational or "therapeutic use" of Cannabis apart from study sessions will be actively discouraged.

E.5 Privacy Issues:
Assigning each study participant an identification number attached to any study data will protect the subjects' privacy. These records will be stored in a locked file in the study safe under Dr. Russo's supervision at WMC. The only people with access to the information will be Dr. Russo, the co-investigators, and involved federal agencies at their discretion.

E.6 Risks vs. Benefits:
As previously discussed, a significant proportion of migraineurs fail to respond to current therapies for their acute migraine. This study hopes to assess the relative risks and benefits of dronabinol and smoked Cannabis in such a group.

F. Vertebrate Animals:
NOT APPLICABLE

Sponsorship:
To date, Dr. Russo has received $4000 over two years from the Multidisciplinary Association for Psychedelic Studies (MAPS) to pursue federal, state and local approval of the study protocol.

G. Literature Cited:
Abel, E.L., Marihuana: The First Twelve Thousand Years, Plenum, New York, 1980.

Agurell, S., Halldin, M., Lindgren, J-E, Ohlsson, A., Widman, M., Gillespie, H. and Hollister, L., "Pharmacokinetics and Metabolism of Delta-1- Tetrahydrocannabinol and Other Cannabinoids with Emphasis on Man," Pharmacol. Rev. 38: 21-43, 1986.

AJPH, "Access to Therapeutic Marijuana/Cannabis," Amer. J. Public Health 86:441-442, 1996.

Artamonov, M.I., "Frozen Tombs of the Scythians," Scientific American 212(5):101-109, 1965.

Auster, F., and Schafer, J., Arzneipflanzen (7. Lieferung), Veb Georg Thieme, Leipzig, 1955.

Barinaga, M., "Pot, Heroin Unlock New Areas for Neuroscience," Science 258:1882-1884, 1992.

Beckman, H., Treatment in General Practice, Saunders, Philadelphia, 1938.

Benet, S., "Early Diffusion and Folk Uses of Hemp." In: V. Rubin (Ed.), Cannabis and Culture, Mouton, The Hague, 1975.

Benetowa, S., Konopie W Wierzeniach I Zwyczajach Ludowych: Le Chanvre dans les Croyances et les Coutumes Populaires, Warsaw, 1936.

Benusan, S.D., "Marihuana Withdrawal Symptoms," Brit. Med. J. 1:112, 1971.

Boulos, L., Medicinal Plants of North Africa, Reference Publications, 1983.

Brunner, T.F., "Marijuana in Ancient Greece and Rome?: the Literary Evidence," J. Psychedel. Drugs 9:221-225, 1977.

Busse, H., "Pflanzenreste in Vorgeschictlichen Geflussen," Zeitschrift f¸r Ethnologie, 1897.

Camp, W.H., "The Antiquity of Hemp as an Economic Plant," J. N.Y, Bot. Gard. 37:110-114, 1936.

Carter, W.E. and Doughty, P.C., "Social and Cultural Aspects of Cannabis Use in Costa Rica," Ann. NY Acad. Sci. 282:2-16, 1976.

Cary, E.H., "Report of Committee on Legislative Activities," JAMA 108:2214, 1937.

Chait, L.D., and Zacny, J.P., "Reinforcing and Subjective Effects of oral Delta-9-THC and Smoked Marijuana in Humans," Psychopharmacology 107:255-262, 1992.

Chiang, C.N. and Barnett, G., "Marijuana Effect and Delta-9- Tetrahydrocannabinol Plasma Level," Clin. Pharm. Therap. 36:234-238, 1984.

Chopra, I.C. and Chopra, R.W., "The Use of Cannabis Drugs in India," Bull. on Narcotics 9:4-29, 1957.

Chopra, R.N., Chopra, I.C., Handa, K.L., and Kapur, L.D., Chopra's Indigenous Drugs of India, Dhur and Sons, Calcutta, 1958.

Co, B.T., Goodwin, D.W., Gado, M., Mikhael, M. and Hill, S.Y., "Absence of Cerebral Atrophy in Chronic Cannabis users by Computerized Transaxial Tomography," JAMA 237:1229-1230, 1977.

Cohen, S., "The 94-Day Cannabis Study," Ann. NY Acad. Sci. 282:211-220, 1976.

Comitas, L., "Cannabis and Work in Jamaica: A Refutation of the Amotivational Syndrome," Ann. NY Acad. Sci. 282:24-32, 1976.

Cowperthwaite, A.C., A Textbook of Materia Medica and Therapeutics, Gross and Delbridge, Chicago, 1891.

Crancer, A., Dille, J.M., Delay, J.C., Wallace, J.E. and Haykin, M.D., "Comparison of the Effects of Marihuana and Alcohol on Simulated Driving Performance," Science 164:851-854, 1969.

Culpeper, N., Culpeper's Complete Herbal, Foulsham and Co., London, undated.

Da Orta, Garcia, Colloquies on the Simples and Drugs of India, Henry Sotheran, London, 1913.

Darmsteter, J. (Trans.), The Zend-Avesta, Part I, The VendÓd‚d, Oxford, London, 1895.

Dastur, J.F., Medicinal Plants of India and Pakistan, Taraporevala, Bombay, undated.

De Selincourt, A. (Trans.), Herodutus: The Histories, Penguin, Baltimore, 1954.

Devane, W.A., Hanus, L., Breuer, A., Pertwee, R.G., Stevenson, L.A., Griffin, G., Gibson, D., Mandelbaum, A., Etinger, A. and Mechoulam, R., "Isolation and Structure of a Brain Constituent That Binds to the Cannabinoid Receptor," Science 258:1946-1949, 1992.

Dinand, U.P., Handbuch der Heilpflanzentunde, Schr. Esslingen, 1921.

Dioscorides, P., Materia Medica.

Dolan, J.P., "A Note on the Use of Cannabis sativa in the 17th Century," J. of the South Carolina Med. Assoc. 67:424-427, 1971.

Dwarakanath, C., "Use of Opium and Cannabis in the Traditional Systems of Medicine in India," Bull. Narcotics 17:15-19, 1965.

El-Mallakh, R.F., "Marijuana and Migraine," Headache 27:442-443, 1987.

Emboden, W.A., "Ritual Use of Cannabis sativa L.: A Historical-Ethnographic Survey." In: P.T. Furst (Ed.): Flesh of the Gods: The Ritual Use of Hallucinogens, Waveland Press, Prospect Heights, 1972.

Emboden, W.A., "The Genus Cannabis and the Correct Use of Taxonomic Categories," J. of Psychoactive Drugs 13:15-21, 1981.

Fan, P., "Cannabinoid Agonists Inhibit the Activation of 5-HT3 Receptors in Rat Nodose Ganglion Neurons," J. of Neurophys. 73:907-910, 1995.

Fantus, B., "Advances in Therapeutic Technic," JAMA 105:877-881, 1933.

Farlow, J.W., "On the Use of Belladonna and Cannabis Indica by the Rectum in Gynecological Practice," Boston Medical and Surgical Journ. 120:507-509, 1889.

Fishbein, M., "Migraine Associated with Menstruation," JAMA 237:326, 1942.

Folstein, M., Folstein, S., McHugh, P.R., "Mini-Mental State: A Practical Method for grading the Cognitive State of the Patient for the Clinician," J. Psychiatr. Res. 12:189-198, 1975.

Foltin, R.W., Fischman, M.W. and Byrne, M.F., "Effects of Smoked Marijuana on Food Intake and Body Weight of Humans Living in a Residential Laboratory," Appetite 11:1-14, 1988.

Fox, R.H., "Headaches: A Study of Some Common Forms with Special Reference to Arterial Tension and to Treatment," Lancet 2:307-309, 1897.

Fride, E. and Mechoulam, R., "Pharmacological Activity of the Cannabinoid Receptor Agonist, Anandamide, a Brain Constituent," Europ. J. Pharmacol. 231:313-314, 1993.

Galen, C., De Facultatibus Alimentorum.

Gieringer, D., "Waterpipe Study," Bull. of Multidisc. Assoc. For Psych. Stud. 6:59-63, 1996.

Gieringer, D., "Why Marijuana Smoke Harm Reduction?" Bull. of Multidisc. Assoc. For Psych. Stud. 6:64-66, 1996.

Goadsby, P.J. and Gundlach, A. L., "Localization of [3H]-Dihydroergotamine Binding Sites in the Cat Central Nervous System: Relevance to Migraine," Ann. Neurol. 29:91, 1991.

Gowers, W.R., A Manual of Diseases of the Nervous System, Blakiston, Philadelphia, 1888.

Grinspoon, L. and Bakalar, J.B., Marihuana: The Forbidden Medicine, Yale Univ., New Haven, 1993.

Grinspoon, L. and Bakalar, J.B., "Marihuana as Medicine: A Plea for Reconsideration," JAMA 273:1875, 1995.

Gunther, R.T., The Greek Herbal of Dioscorides, Hafner, London, 1968.

Hall, W., Solowij, N. and Lemon, J., "The Health and Psychological Consequences of Cannabis Use," National Drug Strategy Monograph Series No. 25, National Drug and Alcohol Research Centre, Australia, 1995.

Hare, H.A., "Clinical and Physiological Notes on the Action of Cannabis Indica, Therapeutic Gazette 2: 225-228, 1887.

Hare, H.A., A Textbook of Practical Therapeutics, Lea and Febiger, Philadelphia, 1922.

Headache Classification Committee of the International Headache Society, "Classification and Diagnostic Criteria for Headache Disorders, Cranial Neuralgias, and Facial Pain," Cephalalgia 8(suppl. 7):1-96, 1988.

Herkenham, M.A., "Localization of Cannabinoid Receptors in Brain: Relationship to Motor and reward Systems," pp. 187-200, in Biological Basis of Substance Abuse, Oxford Univ. Press, New York and London, 1993.

Hollister, L.E., "Health Aspects of Cannabis," Pharmacol. Rev. 38:1-20, 1986.

Humphrey, P.P.A., Feniuk, W. and Perren, M.J., "Anti-Migraine Drugs in Development: Advances in Serotonin Receptor Pharmacology," Headache 20(suppl.):12-16, 1990.

Indian Hemp Drugs Commission. Indian Hemp Drugs Commission Report, Simla, India, 1893-1894.

Johnson, K.M., Dewey, W.L., and Bloom, A.S., "Adrenalectomy Reverse the Effects of Delta-9-THC on Mouse Brain 5-Hydroxytryptamine Turnover," Pharmacology 23:223-229, 1981.

Kabel"k, J., Kreje", Z, and Santavy, F., "Cannabis as a Medicament," Bull. on Narcotics 12(3):5-23, 1960.

Kaempfer, E., Exotic Pleasures: Fascicle III : Curious Scientific and Medical Observations (Library of Renaissance Humanism), Robert W. Carrubba (Translator), S. Illinois Univ. Press, 1996.

Kuehnle, J., Mendelson, J.H., Davis, K.R. and New, P.F.J., "Computed Tomographic Examination of Heavy Marihuana Smokers," JAMA 237:1231- 1232, 1977.

La Guardia Commission, The Marihuana Problem in the City of New York, 1944.

Lewis, B., MÈnage, V.L., Pellat, CH., and Schacht, J., The Encyclopedia of Islam, E.J. Brill, Leiden, 1971.

Lewis, H.E., "Cannabis Indica," JAMA 35:457-458, 1900

Li, Hui-Lin, "An Archaeological and Historical Account of Cannabis in China," Econ. Bot. 28:437-448, 1974.

Lichtman, A.H., Cook, S.A., and Martin, B.R., "Investigation of Brain Sites Mediating Cannabinoid-Induced Antinociception in Rats: evidence Supporting Periaqueductal Gray Involvement," J. Pharmacol. Exper. Therap. 276:585-593, 1996.

Lilly's, Handbook of Pharmacy and Therapeutics, Eli Lilly and Co., Indianapolis, 1898.

Lindgren, J-E., Ohlsson, A., Agurell, S., Hollister, L. and Gilles-Pie, H., "Clinical effects and Plasma Levels of Delta-9-Tetrahydrocannabinol (Delta-9-THC) in Heavy and Light Users of Cannabis," Psychopharmacology 74:208-212, 1980.

Linet, M.S., Stewart, W.F., Celentano, D.D., "An Epidemiological Study of Headache Among Adolescents and Young Adults," JAMA 261:2211- 2216, 1989.

Lipton, R.B., Stewart, W.F., "Migraine in the United States: Epidemiology and Health Care Use," Neurology 43(suppl.):6-10, 1993.

Lozano, I., "El Uso Terapeutico del Cannabis sativa L. en la Medicina ¡rabe," Asclepio 49:199-208, 1997.

Mackenzie, S., "Indian Hemp in Persistent Headache," JAMA 9:732, 1887.

Marshall, C.R., A Text-Book of Materia Medica, Churchill, London, 1905.

Martin, B.R., "Cellular Effects of Cannabinoids," Pharmacol. Rev. 38: 45-74, 1986.

Martin, M.A., "Ethnobotanical Aspects of Cannabis in Southeast Asia." In: V. Rubin (Ed.), Cannabis and Culture, Mouton, The Hague, 1975.

Martin W.J., Hohmann, A.G., and Walker, J.M., "Suppression of Noxious Stimulus-Evoked Activity in the Ventral Posterolateral nucleus of the Thalamus by a Cannabinoid Agonist: Correlation between Electrophysiological and Antinociceptive Effects," J of Neuroscience 16(20):6601-6611, 1996.

Marx, J., "Marijuana Receptor Gene Cloned," Science 249:624-626, 1990.

Mason, A.P. et al., "Cannabis: Pharmacology and Interpretation of Effects," J. Forensic Sci. 30:615-631, 1985.

Mathew, N.T., "Serotonin 1D (5-HT 1D) Agonists and Other Agents in Acute Migraine," Neurologic Clinics 15:61-83, 1997.

Matsuda, L.A., Lolait, S.J., Brownstein, M.J., Young, A.C., and Bonner, T.I., "Structure of a Cannabinoid Receptor and Functional Expression of the Cloned cDNA," Nature 346:561-564, 1990.

Mattison, J.B., "Cannabis Indica as an Anodyne and Hypnotic," St. Louis Med. Surg. Journ. 61:265-271, 1891.

Mauskop, A., Altura, B.T., Cracco, R.Q, and Altura, B.M., "Intravenous Magnesium Sulfate Rapidly Alleviates headaches of Various Types," Headache 36:154-160, 1996.

McConnell, J.F.P., "Uses of Cannabis Indica," The Practitioner 40:95-98, 1888.

Mikuriya, T.H. (ed.), Marijuana: Medical Papers 1839-1972, Medi-Comp Press, Oakland, CA, 1973.

Mikuriya, T.H., "Marijuana in Medicine: Past, Present and Future," Calif. Med. 110:34-40, 1969.

Mikuriya, T.H., "Medicinal Uses of Cannabis at a Buyers' Club," 1995: http://206.61.184.43/schaffer/hemp/sfbc1.htm

Mikuriya, T.H., " Chronic Migraine Headache: Five Cases Successfully Treated with Marinol and/or Illicit Cannabis,"
1997: http://206.61.184.43/schaffer/hemp/migrn1.htm

Milstein, S.L., MacCannell, K., Karr, G. and Clark, S., "Marijuana-Produced Changes in Pain Tolerance: Experienced and Non-Experienced Subjects," Int. Neuropsychiatr. 10:177-182, 1975.

Noyes, R. and Baram, D.A., "Cannabis Analgesia," Compr. Psychiatr. 15:531-535, 1974.

Noyes, R., Brunk, F., Baram, D.A. and Canter, A., "Analgesic Effect of Delta-9-Tetrahydrocannabinol," J. Clin. Pharmacol. 15:134-143, 1975.

Noyes, R., Brunk, F., Avery, D.H. and Canter, A., "The Analgesic Properties of Delta-9-Tetrahydrocannabinol and Codeine," Clin. Pharmacol. And Ther. 18:84-89, 1975.

Ohlsson, A., Lindgren, J-E., Wahlen, A., Agurell, S., Hollister, L.E. and Gillespie, B.A., "Plasma Delta-9-Tetrahydrocannabinol Concentration and Clinical Effects After Oral and Intravenous Administration and Smoking," Clin. Pharmacol.Ther. 28:408-416, 1980.

O'Shaughnessy, W.B., "On the Preparations of the Indian Hemp, or Gunjah (Cannabis Indica); Their Effects on the Animal System in Health, and Their Utility in the Treatment of Tetanus and Other Convulsive Diseases," Trans. Med. And Phys. Soc., Bengal, 71-102, 421-461, 1838-1840.

Osler, W. and McCrae, T., The Principles and Practice of Medicine, Appleton and Co., New York and London, 1915.

Peroutka, S.J., "Developments in 5-Hydroxytryptamine Receptor Pharmacology in Migraine," Neurologic Clinics 8: 829-839, 1990.

Perry, L.M., Medicinal Plants of East and Southeast Asia, MIT Press, Cambridge, 1980.

Ponglux, D., Wongseripipatana, S., et al., Medicinal Plants, Medicinal Plants Exhibition Committee, Bangkok, 1987.
Raskin, N.H., Headache, Churchill Livingstone, New York, 1988.

Ratnam, E.V., "Cannabis Indica," J. Brit. Med. Assoc., Ceylon Branch 13:30-34, 1916.

Remington, J.P. and Wood, H.C., The Dispensatory of the United States of America, Twentieth Edition, Philadelphia, 1918.

Robinson, V., "Historical Notes," Ciba Symposium 8(5-6):401-403, 1946.

Rosner, F., Moses Maimonides' Glossary of Drug Names, American Philosophical Society, Philadelphia, 1979.

Rudenko, S.I., Frozen Tombs of Siberia: The Pazyryk Burials of Iron Age Horsemen, Univ. of California, Berkeley, 1970.

Russell, J.R., "Therapeutical Uses and Toxic Effects of Cannabis Indica," Lancet 1:637-638, 1890.

Russo, E.B., "Headache Treatments by Native Peoples of the Ecuadorian Amazon: A Preliminary Cross-Disciplinary Assessment," J. Ethnopharmacology 36: 193-206, 1992.

Russo, E.B., "Ecuador Rain Forest International Headache Project," book chapter in Reed, D.R. (ed.), Spirit of Enterprise: The 1993 Rolex Awards, Buri International, Bern, Switzerland, 1993.

Russo, E.B., Medora, R., Parker, K. and Thompson, C., "Schedule 1 Research Protocol: An Investigation of Psychedelic Plants and Compounds for Activity in Serotonin Receptor Assays for Headache Treatment and Prophylaxis," Bull. Multidisc. Assoc. Psych. Stud. 7:4-8, 1997.

Russo, E.B., "Cannabis for Migraine Treatment: The Once and Future Prescription?: An Historical and Scientific Review," Pain 76(1):3-8, 1998

Schultes, R.E., "Man and Marijuana," Nat. Hist. 82:59-63, 80, 82, 1973.

Schultes, R.E. and Hofmann, A., The Botany and Chemistry of the Hallucinogens, Thomas, Springfield, IL, 1980.

Schultes, R.E., Klein, W.M., Plowman, T. and Lockwood, T.E., "Cannabis: An Example of Taxonomic Neglect," Bot. Mus. Leafl. Harvard Univ. 23:337-367, 1974.

Sellman, D., "Safe Use of Cannabis," New Zealand Med. J. 105:531, 1993.

Sidney, S., Beck, J.E., Tekawa, I.S., Quesenberry, C.P., and Friedman, G.D., "Marijuana Use and Mortalilty," Am. J. Publ. Health 87:585-590, 1997.

Spadone, C., "Neurophysiologie du Cannabis," EncÈphale 17:17-22, 1991.

Stevens, A.A., The Practice of Medicine, Saunders, Philadelphia, 1926.

Stewart, W.F., Lipton, R.B., Celantano, D.D., "Prevalence of Migraine Headache in the United States," JAMA 267:64-69, 1992.

Suckling, C.W., "On the Therapeutic Value of Indian Hemp," Brit. Med. J. 2:12, 1891.

Volfe, Z., Dvilansky, A., and Nathan, I., "Cannabinoids Block Release of Serotonin from Platelets by Plasma from Migraine Patients," Int. J. Clin. Pharm. Res. 4:243-246, 1985.

Voth, E.A., and Schwartz, R.H., "Medical Applications of Delta-9- Tetrahydrocannabinol and Marijuana," Ann. Int. Med. 126:791-798, 1997.

Wall, M.E., Sadler, B.M., Brine, D., Taylor, H. and Perez-Reyes, M., "Metabolism, Disposition, and Kinetics of Delta-9-Tetrahdrocannabinol in Men and Women," Clin. Pharm. Therap. 34:352-362, 1983.

Walton, R.P., Marihuana: Americas's New Drug Problem: A Sociologic Question with its Basic Explanation Dependent on Biologic and Medical Principles, J.B. Lippincott, Philadelphia, 1938.

Waring, E.J., Practical Therapeutics, Lindsay and Blackiston, Philadelphia, 1874.

Weber, J.T., O'Connor, M-F, Hayakataka, K., Colson, N., Medora, R., Russo, E.B., and Parker, K.K., "Activity of Parthenolide at 5HT2A Receptors," J. of Natl. Prod. 60:651-653, 1997.

Weil, A.T., Zinberg, N.E. and Nelsen, J.M., "Clinical and Psychological Effects of Marihuana in Man," Science 162:1234-1242, 1968.

Young, F.L., "In the Matter of Marijuana Rescheduling Petition," Dept. of Justice, Drug Enforcement Administration. Docket 86-22. Washington, D.C.: Drug Enforcement Administration, September 6, 1988.

Zias, J., Stark, H., Seligman, J. et al., "Early Medical Use of Cannabis," Nature 363:215, 1993.

Zimmer, L., and Morgan., J.P., Marijuana Myths Marijuana Facts, Lindesmith Center, New York, 1997.

H. Consortium/Contractual Arrangements:
NOT APPLICABLE

 


 

INTERVIEW PROTOCOL

1. Have you been formally diagnosed with migraines?     Yes     No

    How often do you get headaches?

2. Where is the pain located?

 

3. Is it worse on one side than the other?     Yes      No

4. Do other family members have headaches?     Yes      No

5. If so, what relationship are they?

 

6. Did you feel the need to lie on the couch with a cool damp cloth on your head to relieve     headache?      Yes &nbs p;   No

7. Is there a beating quality to the pain?     Yes      No

8. If you climb stairs, exert yourself, or bend over when you have a headache, is there a beating     quality?      Yes      No

9. When you have a headache do you feel sick to your stomach, lose your appetite, or      vomit?      Yes     No

10. Or, when you have a headache, are you able to sit up to a full meal?     Yes      No

11. When you have a headache do you experience any visual problems?     Yes      No

12. If so, is there blurring, or do you see spots or sparkles?     (circle which ones)

13. Are your eyes sensitive to light?     Yes      No

14. Do you prefer to be in a dark room?     Yes      No

15. Can you tolerate bright sunlight during a headache?     Yes      No

16. Does loud music bother you then?     Yes      No

17. Do certain sounds bother you?     Yes      No

18. Are you currently under a doctor's care?     Yes      No

19. Have you had cardiac problems in the past or currently?     Yes      No

20. Are you currently pregnant or breast- feeding?     Yes      No

21. Have you ever been diagnosed with a psychiatric diagnosis?     Yes      No

      If so, list diagnosis

 

22. Has anyone in your family been diagnosed with a psychiatric disorder?     Yes     No

      If so, list diagnosis

23. Are you currently taking any of the following types of medication: antidepressants, anticonvulsants       (i.e., seizure medication), or tranquilizers?     Yes      No

24. Have you used hallucinogens or opiates [e.g., LSD, Mescaline, Peyote, STP, DMT, Psilocybin       (mushrooms), Heroin, Morphine, Opium]?     Yes     No

25. Have you used marijuana recreationally?     Yes       No

26. Have you used crack, cocaine, or Ecstasy?     Yes     No

27. Have you used inhalants?     Yes     No

28. Have you used stimulants?     Yes     No

29. Have you used anti-anxiety agents or sleeping medications?     Yes     No

30. Have you used pain medication?     Yes     No

31. Have you been treated for alcoholism?     Yes     No

32. Are you taking any medications not already listed?     Yes     No

33. This treatment involves the use of Dronabinol (synthetic THC), meperidine (Demerol) and Cannabis       (marijuana). Are you willing to use this medication as part of this research, recognizing its       government approval and legality for this study?     Yes     No

34. This treatment requires that you be accompanied to and from treatment by a licensed driver who will       be able to escort you home.

      Is someone available and willing to do this for you?     Yes     No

35. Is your job subject to drug testing?     Yes     No

36. Have you received sumatriptan injections for your migraines?     Yes     No

37. How many times?

 

38. Was it effective for the pain?     Yes     No

39. Was it effective for the nausea?     Yes     No

40. Was it effective for photophobia?     Yes     No

41. Did sumatriptan give you side effects?     Yes     No

42. If so, what were they?

 

43. Would you want to have sumatriptan injections again for your headaches?     Yes     No

44. Why, or why not?


 

Subject #
Treatment #
Date
Current Time

Functioning Questionnaire: 1 Hour Post Treatment

Please complete the following questions as carefully as you can, and return to the study nurse.

1. Do you have a job?     Yes     No

A. If yes, could you work with the way you felt immediately following treatment?     Yes     No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate).

     0%   &nbp;  &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

B. If yes, could you work with the way you feel right now?     Yes     No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate)

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%       90%   ;     100%

2. Are you a student?     Yes     No

A. If yes, could you study with the way you felt immediately following treatment?     Yes     No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate).

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%       90%   ;     100%

B. If yes, could you study with the way you feel right now?     Yes     No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate).

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%       90%   ;     100%

C. If yes, could you have gotten anything out of class with the way you felt right after the      treatment?     Yes&nbs p;    No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate).

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%       90%   ;     100%

D. If yes, could you have gotten anything out of class with the way you feel right now?     Yes     No

     How effective would you be? (Please put a slash at the percentage of effectiveness you would      anticipate).

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%       90%   ;     100%

3. How would you rate your pain right now? (Please put a slash at the number that describes your     current level of pain where 0 is no pain and 10 is intense pain).

       0     &n bsp; 1       2   &nbs p;   3       4        5       6& nbsp;      7     &nbs p; 8       9        10

No pain                         &nb sp;          Neutral & nbsp;             ;            &nb sp;     Intense pain

4. How would you rate your nausea right now? (Please put a slash at the number that describes your     current level of nausea where 0 is no nausea and 10 is severe nausea).

       0     &n bsp; 1       2   &nbs p;   3       4        5       6& nbsp;      7     &nbs p; 8       9        10

No nausea           &nbs p;            &n bsp;      Neutral                  &nb sp;            & nbsp;Severe nausea

5. How would you rate your sensitivity to light? (Please put a slash at the number that describes your     current level of sensitivity where 0 is none and 10 is extremely sensitive).

       0     &n bsp; 1       2   &nbs p;   3       4        5       6& nbsp;      7     &nbs p; 8       9        10

Not sensitive           &n bsp;                Neutral        &nb sp;            & nbsp;      Extremely sensitive

6. How would you rate your sensitivity to sound? (Please put a slash at the number that describes your     current level of sensitivity where 0 is none and 10 is extremely sensitive).

       0        1       2   & nbsp;   3       4 &nb sp;     5        6       7     & nbsp; 8       9   &nb sp;   10

Not sensitive           &n bsp;                Neutral        &nb sp;            & nbsp;       Extremely sensitive

7. Please describe any negative experiences you had during or after the treatment.

 

8. What medicine would you say that you received?

 


 

Subject #
Treatment #
Date of Treatment

Functioning Questionnaire- 2 hours Post Treatment

Please complete the following questions as carefully as you can, and return them to the study nurse.

1. What time is it right now?

2. Do you have a job?     Yes     No

    If yes, could you work with the way you felt 2 hours after treatment?     Yes     No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

3. Are you a student?     Yes     No

    If yes, could you study with the way you feel 2 hours after treatment?     Yes     No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

B. If yes, could you have gotten anything out of class with the way you feel 2 hours after     treatment?     Yes &nbs p;   No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

4. How would you rate your pain 2 hours after treatment? (Please put a slash at the number that     describes your level of pain where 0 is no pain and 10 is severe pain).

     0       1& nbsp;      2     &nbs p; 3       4        5       6  & nbsp;    7       8&nb sp;      9        10

No pain                         &nb sp;       Neutral    & nbsp;             ;            &nb sp;   Severe pain

5. How would you rate your nausea 2 hours after treatment? (Please put a slash at the number that     describes your current level of nausea where 0 is no nausea and 10 is severe nausea).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No nausea           &nbs p;            &n bsp;   Neutral                     &nb sp;          Severe nausea

6. How would you rate your sensitivity to light 2 hours after treatment? (Please put a slash at the number     that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;             Neutral           &nb sp;            & nbsp;   Extremely sensitive

7. How would you rate your sensitivity to sound 2 hours after treatment? (Please put a slash at the     number that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;             Neutral           &nb sp;            & nbsp;   Extremely sensitive

8. Please describe any negative experiences you had since the treatment.

 

9. What medicine would you say that you received?

 


 

Subject #
Treatment #
Date of Treatment

Functioning Questionnaire- 3 hours Post Treatment

Please complete the following questions as carefully as you can 3 hours after treatment, and return it to the study nurse.

1. What time is it right now?

2. Do you have a job?     Yes     No

    If yes, could you work with the way you feel 3 hours after treatment?     Yes     No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

3. Are you a student?     Yes     No

   A. If yes, could you study with the way you feel 3 hours after treatment?     Yes     No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

   B. If yes, could you have gotten anything out of class with the way you feel 3 hours after    treatment?     Yes  &nbs p;  No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

4. How would you rate your pain 3 hours after treatment? (Please put a slash at the number that     describes your level of pain where 0 is no pain and 10 is severe pain).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No pain                         &nb sp;       Neutral    & nbsp;             ;            &nb sp;   Severe pain

5. How would you rate your nausea 3 hours after treatment? (Please put a slash at the number that     describes your current level of nausea where 0 is no nausea and 10 is severe nausea).

     0       1& nbsp;      2     &nbs p; 3       4        5       6  & nbsp;    7       8&nb sp;      9        10

No nausea           &nbs p;            &n bsp;   Neutral                     &nb sp;           Severe nausea

6. How would you rate your sensitivity to light 3 hours after treatment? (Please put a slash at the number     that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;    Extremely sensitive

7. How would you rate your sensitivity to sound 3 hours after treatment? (Please put a slash at the     number that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;   Extremely sensitive

8. Please describe any negative experiences you have had since the treatment.

 

9. What medicine would you say that you received?

 

10. Do you need the rescue medication?

 


 

Subject #
Treatment #
Date of Treatment

Functioning Questionnaire- 4 hours Post Treatment

Please complete the following questions as carefully as you can 4 hours after treatment, and return it to the study nurse.

1. What time is it right now?

2. Do you have a job?     Yes     No

    If yes, could you work with the way you feel 4 hours after treatment?     Yes     No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

3. Are you a student?     Yes     No

    A. If yes, could you study with the way you feel 4 hours after treatment?     Yes     No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

B. If yes, could you have gotten anything out of class with the way you feel 4 hours after     treatment?     Yes &nbs p;   No

    How effective would you be? (Please put a slash at the percentage of effectiveness you would     anticipate).

     0%       1 0%       20%    &nb sp;  30%       40%        50%        60%       70%    &n bsp;  80%        90%  ;      100%

4. How would you rate your pain 4 hours after treatment? (Please put a slash at the number that     describes your level of pain where 0 is no pain and 10 is severe pain).

     0       1& nbsp;      2     &nbs p; 3       4        5       6  & nbsp;    7       8&nb sp;      9        10

No pain                         &nb sp;        Neutral   & nbsp;             ;            &nb sp;    Severe pain

5. How would you rate your nausea 4 hours after treatment? (Please put a slash at the number that     describes your current level of nausea where 0 is no nausea and 10 is severe nausea).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No nausea           &nbs p;            &n bsp;   Neutral                     &nb sp;          Severe nausea

6. How would you rate your sensitivity to light 4 hours after treatment? (Please put a slash at the number     that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;     Extremely sensitive

7. How would you rate your sensitivity to sound 4 hours after treatment? (Please put a slash at the     number that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;    Extremely sensitive

8. Please describe any negative experiences you have had since the treatment.

 

9. What medicine would you say that you received?

 

10.Did you receive the rescue medication?     YES______    &n bsp;NO_____

11. If so, what effects has it had?

 


 

Subject #
Treatment #
Date of Treatment

Functioning Questionnaire- 24 hours Post Treatment

Please complete the following questions as carefully as you can 24 hours after treatment. Then return them in the self-addressed, stamped envelope provided.

1. What time is it right now?

2. Do you have a job?     Yes     No

   If yes, can you work with the way you feel 24 hours after treatment?     Yes     No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

3. Are you a student?     Yes     No

   A. If yes, can you study with the way you feel 24 hours after treatment?     Yes     No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%        0%     & nbsp; 20%       30%  &nbs p;    40%       50%& nbsp;      60%     & nbsp; 70%       80%  &nbs p;    90%       100%

   B. If yes, can you have get anything out of class with the way you feel 24 hours after    treatment?     Yes  &nbs p;  No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

4. How would you rate your pain 24 hours after treatment? (Please put a slash at the number that     describes your level of pain where 0 is no pain and 10 is severe pain).

     0       1& nbsp;      2     &nbs p; 3       4        5       6  & nbsp;    7       8&nb sp;      9        10

No pain                         &nb sp;       Neutral    & nbsp;             ;            &nb sp;   Severe pain

5. How would you rate your nausea 24 hours after treatment? (Please put a slash at the number that     describes your current level of nausea where 0 is no nausea and 10 is severe nausea).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No nausea           &nbs p;            &n bsp;   Neutral                     &nb sp;           Severe nausea

6. How would you rate your sensitivity to light 24 hours after treatment? (Please put a slash at the     number that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;    Extremely sensitive

7. How would you rate your sensitivity to sound 24 hours after treatment? (Please put a slash at the     number that describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;              Neutral          &nb sp;            & nbsp;    Extremely sensitive

8. Please describe any negative experiences you have had since the treatment.

 

9. What medicine would you say that you received in the session?

 


 

Subject #
Treatment #
Date of Treatment

Pre-Treatment Questionnaire

1. How many headaches have you had since the last treatment?

2. How satisfied were you with the last treatment? (Circle the appropriate number).

     1        2       3     & nbsp; 4       5   &nb sp;   6       7   ;     8       9& nbsp;      10

Not satisfied           &n bsp;     Neutral                   &nb sp;          Extremely Satisfied

3. Please write any comments that you would like to share (i.e., problems, concerns, and likes/dislikes).

 

4. Has your medication use changed at all (type, frequency of use) since your last study treatment?

 

5. Please write any comments that you would like to share (i.e., problems, concerns, and likes/dislikes).

 

6. What time is it right now?

7. Do you have a job? Yes No

   If yes, can you work with the way you feel right now?     Yes     No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

8. Are you a student?     Yes     No

   A. If yes, can you study with the way you feel right now?     Yes     No

   How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

   If yes, could you get anything out of class with the way you feel right now?     Yes     No

   C. How effective would you be? (Please put a slash at the percentage of effectiveness you would    anticipate).

     0%      &nbs p;10%       20%    & nbsp;  30%       40% &nbs p;     50%      &nbs p;60%       70%    & nbsp;  80%       90% &nbs p;     100%

9. How would you rate your pain right now? (Please put a slash at the number that describes your level     of pain where 0 is no pain and 10 is severe pain).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No pain                         &nb sp;        Neutral   & nbsp;             ;            &nb sp;    Severe pain

10. How would you rate your nausea right now? (Please put a slash at the number that describes your       current level of nausea where 0 is no nausea and 10 is severe nausea).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

No nausea           &nbs p;            &n bsp;   Neutral                     &nb sp;           Severe nausea

11. How would you rate your sensitivity to light right now? (Please put a slash at the number that       describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;             Neutral           &nb sp;            & nbsp;    Extremely sensitive

12. How would you rate your sensitivity to sound right now? (Please put a slash at the number that       describes your current level of sensitivity where 0 is none and 10 is extremely sensitive).

     0        1       2     & nbsp; 3       4   &nb sp;   5       6   ;     7       8& nbsp;      9     &nbs p; 10

Not sensitive           &n bsp;             Neutral           &nb sp;            & nbsp;    Extremely sensitive

 


 

Statement of Informed Consent

Pilot Study: Inhalation of Pyrolysed Cannabis sativa vs. Oral Dronabinol, and Injected Sumatriptan as Treatment for Acute Migraine: A Prospective Double-blind Crossover Study

Principal Investigator: Ethan Russo, M.D.
Co-Investigators: Stuart Hall, Ph.D.; Jeannine Mielke; Laura Taylor Painter,
Psychology Dept., University of Montana; Brian Steele, Ph.D., Mathematics Dept.


Before taking part in this study, you need to be informed of the following information to allow you to consent to your participation in this project. Indicate your consent by signing at the bottom:

The purpose of this study is to enhance knowledge of, and to examine and compare the efficacy of, three different drug treatments and two placebo treatments for migraines. These include:

Dronabinol (Marinol, synthetic THC, the active ingredient of marijuana).
A placebo capsule (a placebo contains no active medicine).
Inhaled smoked Cannabis (marijuana).
Placebo (inactive) Cannabis containing no THC.
Injected sumatriptan (Imitrex). Patients who have had bad side effects with injected sumatriptan, but no true allergic reaction may have the option of receiving intra-nasal sumatriptan.

This project is important because there is a need for new medications that will effectively treat acute migraines. Some patients and their doctors have claimed that smoked Cannabis (marijuana) is helpful in treating migraines, and we would like to test that idea experimentally. Alternative medications and non-drug treatments for migraine exist in addition to the study medicines. Questions about alternative treatments should be directed to your regular physician or care provider.
This project has been approved by the Food and Drug Administration (FDA), Drug Enforcement Agency (DEA), National Institute of Health, National Institute on Drug Abuse (NIDA), and the St. Patrick Hospital/Community Hospital Joint Investigational Review Board (IRB). It will be considered a legal experimental use of marijuana (Cannabis) for this research project.

As one of about 40 participants in this study, you will be administered each of these treatments by Dr. Ethan Russo, a neurologist, or research nurses during the study period of two three month blocks with a two week break in between. Each subject will be treated with injected sumatriptan, or will receive a capsule and smoke a special cigarette. Both subjects and administrators will be "blinded." That is a way of saying that neither side will know whether it is true marijuana or dronabinol that is received as opposed to placebo. No one will receive both true marijuana and true dronabinol in a single session, and no one will receive only two placebos in a session. Two blood samples will be taken during each session, with the exception of the sumatriptan sessions. You will also be asked to respond to oral and written questions concerning your impressions of the treatments during four hours. If you are still feeling badly after three hours, you will have the option of receiving "rescue medication," in the form of an injection of magnesium sulfate. After four hours, you may return home with your "designated driver," or via taxi.
You will be asked to fill out and return a questionnaire the day after your treatment. The researchers may contact you by telephone if any further information is needed.
The questionnaires and interview data will be reviewed by Dr. Russo, the faculty researcher associated with the projects, and two trained research assistants who are graduate students in psychology at the University of Montana. These individuals will be committed to confidentiality. This confidentiality will be maintained throughout this process by assigning a code number to your records, which will be used throughout the data analysis procedures. Your identity will not be divulged to anyone else, but the IRB and federal agencies involved may wish to review your record with the code number as the identifier.
Records will be locked in a safe.

There are several potential benefits of this research. The data we collect will provide information about the effects of these drug treatments on migraines and about patients' ability to function following each treatment. This data may or may not help to make migraine treatment more effective for you and other patients in the future.

Potential risks of the study may include decreased ability to function after a drug treatment, physical discomfort from injections of sumatriptan or blood drawing. Some people become anxious after exposure to THC from dronabinol or Cannabis, especially if they have not previously experienced it. However, this side effect is usually brief, and we will make efforts to have you as comfortable as possible during the testing procedures.
You will be asked to confine your medicines after a session to your regular prescriptions or ibuprofen or naproxen, if at all possible. If any disturbing side effects are experienced after an experimental session, please contact Dr. Russo or the study nurses. Failing success there, the on-call neurologist can be reached after hours by calling (406)721-5600.
Additionally, it would be important that use of Cannabis (marijuana) during the study period be confined to experimental sessions.
Pregnant women will be excluded from the study. We would also strongly prefer that any women of child-bearing potential be using an effective method of birth control during the study.
Risks involved in this study are minimal. However, the University of Montana requests that the following clause be included, in all consent forms for studies with human participants: "In the event that a participant is physically injured during the course of this research, he or she should individually seek appropriate medical treatment. If the injury is caused by the negligence of the University or any of its employees, the participant may be entitled to reimbursement or compensation pursuant to the Comprehensive State Insurance Plan established by the Department of the Administration under the authority of the M.C.A., Title 2, Chapter 9. In the event of a claim for such personal injury, further documentation may be obtained from University Legal Counsel."

In the unlikely event of unforeseen complications or injury, appropriate medical intervention will be arranged for you, and billed through your insurance carrier, if possible. The researchers will not be able to provide any other compensation to you if you are injured.

Monetary reimbursement of $30 will be provided for your effort for each of the treatment sessions that take place during data collection. There will be no cost to you for the medications or the visits with Dr. Russo.

The participants of this project may choose to end the session(s) at any time, for any reason without prejudice, but we may follow up to invite you to rejoin the study. Monetary reimbursement for treatment sessions you begin, but do not complete, will still be guaranteed.

You may be withdrawn from this study without your consent by the investigators for just cause, and this will not be subject to appeal. We hope that this will not become necessary.

Because of the sensitive political nature of this investigation, it is our preference that suitable discretion be employed by participants, and not discussed with other than appropriate individuals prior to its completion.

If the participants have questions or concerns about any aspect of this project, please feel free to contact the researchers, Jeannine B. Mielke and Laura Taylor Painter; the faculty supervisor, Stuart Hall, Ph.D., or the principal investigator, Ethan Russo, M.D.:

Laura T. Painter/Jeannine B. Mielke
Department of Psychology
University of Montana
Missoula, MT 59812
(406) 243-4523

Ethan Russo, M.D.
Department of Neurosciences
Western Montana Clinic
515 West Front St.
Missoula, MT 59802
(406) 721-5600 ext. 7243


Dr. Stuart Hall
Department of Psychology
University of Montana
Missoula, MT 59812
(406) 243-4521


For any questions on your rights as an experimental subject, please contact the IRB chairman, Jim Gouaux, M.D., or the IRB coordinator, Ms. Lola Goss:

Jim Gouaux, M.D./ Ms. Lola Goss
St. Patrick Hospital
500 West Broadway
Missoula, MT 59802
(406) 329-5669


I have read the preceding information and understand it. I also consent to my participation in this project.

Signature:_______________Date:__________


Witness:_________________Date:__________

Please include your name and address if you wish to receive a summary of the research findings when this study is complete. The expected completion date is July 2000.

Name:________________

Address:_____________


This study is being sponsored by the National Institutes of Health (NIH), a Federal Agency in Rockville, MD.


Ethan B. Russo, M.D. __________ Date: _______________

SAMPLE ADVERTISEMENT

The following is a sample of the advertisement we are likely to employ:

MIGRAINE RESEARCH: CALL FOR PATIENTS

"We are looking for individuals to participate in a NIH-approved research project examining the effectiveness of experimental medications in the treatment of migraine. Participants must be over 18, and have three or more severe headaches per month. Treatment with this experimental protocol will be provided free of charge for six months. Monetary reimbursement will be available. Contact the researchers at (telephone number)."


Return to the MAPS Home Page