Find information on thousands of medical conditions and prescription drugs.

Cerebral cavernous malformations

more...

Home
Diseases
A
B
C
Angioedema
C syndrome
Cacophobia
Café au lait spot
Calcinosis cutis
Calculi
Campylobacter
Canavan leukodystrophy
Cancer
Candidiasis
Canga's bead symptom
Canine distemper
Carcinoid syndrome
Carcinoma, squamous cell
Carcinophobia
Cardiac arrest
Cardiofaciocutaneous...
Cardiomyopathy
Cardiophobia
Cardiospasm
Carnitine transporter...
Carnitine-acylcarnitine...
Caroli disease
Carotenemia
Carpal tunnel syndrome
Carpenter syndrome
Cartilage-hair hypoplasia
Castleman's disease
Cat-scratch disease
CATCH 22 syndrome
Causalgia
Cayler syndrome
CCHS
CDG syndrome
CDG syndrome type 1A
Celiac sprue
Cenani Lenz syndactylism
Ceramidase deficiency
Cerebellar ataxia
Cerebellar hypoplasia
Cerebral amyloid angiopathy
Cerebral aneurysm
Cerebral cavernous...
Cerebral gigantism
Cerebral palsy
Cerebral thrombosis
Ceroid lipofuscinois,...
Cervical cancer
Chagas disease
Chalazion
Chancroid
Charcot disease
Charcot-Marie-Tooth disease
CHARGE Association
Chediak-Higashi syndrome
Chemodectoma
Cherubism
Chickenpox
Chikungunya
Childhood disintegrative...
Chionophobia
Chlamydia
Chlamydia trachomatis
Cholangiocarcinoma
Cholecystitis
Cholelithiasis
Cholera
Cholestasis
Cholesterol pneumonia
Chondrocalcinosis
Chondrodystrophy
Chondromalacia
Chondrosarcoma
Chorea (disease)
Chorea acanthocytosis
Choriocarcinoma
Chorioretinitis
Choroid plexus cyst
Christmas disease
Chromhidrosis
Chromophobia
Chromosome 15q, partial...
Chromosome 15q, trisomy
Chromosome 22,...
Chronic fatigue immune...
Chronic fatigue syndrome
Chronic granulomatous...
Chronic lymphocytic leukemia
Chronic myelogenous leukemia
Chronic obstructive...
Chronic renal failure
Churg-Strauss syndrome
Ciguatera fish poisoning
Cinchonism
Citrullinemia
Cleft lip
Cleft palate
Climacophobia
Clinophobia
Cloacal exstrophy
Clubfoot
Cluster headache
Coccidioidomycosis
Cockayne's syndrome
Coffin-Lowry syndrome
Colitis
Color blindness
Colorado tick fever
Combined hyperlipidemia,...
Common cold
Common variable...
Compartment syndrome
Conductive hearing loss
Condyloma
Condyloma acuminatum
Cone dystrophy
Congenital adrenal...
Congenital afibrinogenemia
Congenital diaphragmatic...
Congenital erythropoietic...
Congenital facial diplegia
Congenital hypothyroidism
Congenital ichthyosis
Congenital syphilis
Congenital toxoplasmosis
Congestive heart disease
Conjunctivitis
Conn's syndrome
Constitutional growth delay
Conversion disorder
Coprophobia
Coproporhyria
Cor pulmonale
Cor triatriatum
Cornelia de Lange syndrome
Coronary heart disease
Cortical dysplasia
Corticobasal degeneration
Costello syndrome
Costochondritis
Cowpox
Craniodiaphyseal dysplasia
Craniofacial dysostosis
Craniostenosis
Craniosynostosis
CREST syndrome
Cretinism
Creutzfeldt-Jakob disease
Cri du chat
Cri du chat
Crohn's disease
Croup
Crouzon syndrome
Crouzonodermoskeletal...
Crow-Fukase syndrome
Cryoglobulinemia
Cryophobia
Cryptococcosis
Crystallophobia
Cushing's syndrome
Cutaneous larva migrans
Cutis verticis gyrata
Cyclic neutropenia
Cyclic vomiting syndrome
Cystic fibrosis
Cystinosis
Cystinuria
Cytomegalovirus
Dilated cardiomyopathy
Hypertrophic cardiomyopathy
Restrictive cardiomyopathy
D
E
F
G
H
I
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z
Medicines

Incidence of Occurrence and Symptoms

Cerebral cavernous malformation (CCM), also known as cavernous angioma, cavernous haemangioma, and cavernoma, is a vascular disorder of the central nervous system that may appear either sporadically or exhibit autosomal dominant inheritance. The incidence in the general population is between 0.1-0.5%, and clinical symptoms typically appear between 30 to 50 years of age. Once thought to be strictly congenital, these vascular lesions have been found to occur de novo.

This disease is characterized by grossly dilated blood vessels with a single layer of endothelium and an absence of neuronal tissue within the lesions. These thinly-walled vessels resemble sinusoidal cavities filled with stagnant blood. Blood vessels in patients with CCM can range from a few millimeters to several centimeters in diameter. CCM lesions commonly resemble raspberries in external structure.

Many patients live their whole life without knowing they have a cerebral cavernous malformation. Other patients can have severe symptoms like seizures, headaches, paralysis, bleeding in the brain (cerebral hemorrhage), and even death. The nature and severity severity of the symptoms depend on the lesion's location in the brain. Approximately 70% of these lesions occur in the supratentorial region of the brain; the remaining 30% occur in the infratentorial region.

Symptoms and Diagnosis

Clinical symptoms of this disease include recurrent headaches, focal neurological deficits, hemorrahagic stroke, and seizures, but CCM can also be asymptomatic. Diagnosis is most commonly made by magnetic resonance imaging MRI, but not all MRI exams are created equal. It's paramount that the patient request a gradient-echo MRI (aka T2-Flair) in order to unmask small or punctate lesions which may otherwise remain undetected. Sometimes quiescent CCMs can be revealed as incidental findings during MRI exams ordered for other reasons.

Sometimes the lesion appearance imaged by MRI remains inconclusive. Consequently neurosurgeons will order a cerebral angiogram or magnetic resonance angiogram (MRA). Since CCMs are low flow lesions (they are hooked into the venous side of the circulatory system), they will be angiographically occult (invisible). If a lesion is discernable via angiogram in the same location as in the MRI, then an arteriovenous malformation (AVM) becomes the primary concern.

CCMs & Venous Malformations

Many times a CCM is accompanied by a venous malformation (VM) which are also known as a developmental venous anomaly (DVA). These lesions appear either as enhancing linear blood vessels or caput medusae--a radial orientation of small vessels that resemble the hair of Medusa from Greek Mythology. These are normally benign lesions that provide normal brain drainage into the venous system. Conventional wisdom recommends leaving these lesions alone, even if surgical removal of the CCM is advocated.

Read more at Wikipedia.org


[List your site here Free!]


Guest editorial
From Neurological Research, 1/1/02 by Fessler, Richard D

In less than a lifetime, the treatment of neurovascular disease has progressed at a dizzying pace. Much of this progress has occurred in the last 40 years as technology, improved critical care, and subspecialization have resulted in continuous improvement in patient outcome. Many of the individual practitioners and scientists in the disciplines of neurology, neurosurgery, and radiology who have been responsible for advancing the field are still with us today. A number of us are privileged to count them as our mentors and colleagues. Our ability to treat complex neurovascular disease on a daily basis is due in large part to the efforts of those who struggled before us and have shared their passion, wisdom, and failings with us. In neurosurgery, these recent pioneers include Drake, Yasargil, Sugita, and a bevy of colleagues too numerous to list. Their contributions continue to influence the daily care of neurosurgical patients the world over. Likewise, neuroendovascular surgery owes much of its current status to Serbinenko, Hieshima, Berenstein, Djindjian, Merland, Moret, and Guglielmi. The contributions in devices, techniques, and experience influence the daily practice of endovascular therapy. Many of the 'pioneers' of endovascular therapy are working today and continuing to define the field.

The tremendous clinical successes of neuroendovascular techniques have led to an increase in the number of practitioners with a substantial number coming from specialties outside radiology. Best estimates worldwide suggest that nearly 50% of practitioners in neuroendovascular surgery come from specialties outside radiology, predominantly neurosurgery and neurology. interventional radiologists and neurosurgeons in North America have recognized this growing hybrid subspecialty through the establishment of joint guidelines for the training of neuroendovascular specialists1.

Historically, the first description of a vascular anomaly was an aneurysm from an autopsy case report by the British physician Gilbert Blane in 1800(2). The pre-morbid diagnosis of intracranial aneurysms did not occur until the introduction of lumbar puncture by Quincke in 1891(3). Egas Moniz, the father of modern cerebral angiography, introduced the technique in 1927 for the localization of tumors(4,5). In spite of the relatively recent introduction of methods for identifying vascular lesions, the first successful treatment of an aneurysm was in 1808 by Cooper who performed carotid ligation6. Zeller performed the first direct approach to a carotid cavernous fistula in 1911 and Dott successfully wrapped an intracranial aneurysm in 1931 7,8. The first aneurysm clipping was performed by Dandy in 1937 with a silver clip9. The subsequent introduction of the operating microscope and the development of aneurysm clips in their modern form brought neurovascular surgery into the modern era. One of the first technological improvements in aneurysm clips was Mayfield's introduction of clips that could be reapplied. Heifetz later added a reliable spring mechanism, and Yasargil and Sugita improved upon the early clips with designs that have become the standard the world over10,11. Modern neurosurgery subsequently trained a generation of neurosurgeons in microsurgical techniques12,13.

The origins of endovascular therapy can be traced to the first attempt to induce thrombosis of an aneurysm as reported in 1831 and 1832 by Velpeu and Phillips, respectively 14,15. Each independently placed needles within aneurysms to induce thrombosis. In 1941, Werner and colleagues reported treatment of an aneurysm by filling it with yards of silver wire through a spinal needle16. Mullen was one of the early neurosurgical pioneers in endovascular techniques. Mullen and coworkers induced electrothrombosis in aneurysms by the introduction of an electrode into aneurysms17. The first successful catheterization of intracranial vessels was reported by Lussenhop and Velasquez. Latex detachable balloons were created by Serbinenko, allowing the endovascular occlusion of vessels. The concept of magnetic navigation of microcatheters within the intracranial circulation was introduced by a group led by Askenasy. They prognosticated that some day a magnetically driven catheter would be used in the treatment of aneurysms18-20. This was subsequently expanded upon by Alksne and colleagues as well as Hilal21,22. This pioneering work was the harbinger of the current research involving magnetic field direction of catheters for interventional procedures.

Technical advances in endovascular therapy have moved the field rapidly in a few short years. Guglielmi and his colleagues from UCLA first reported their clinical experience using the technique of electrothrombosis and detachable coils to treat a carotid cavernous fistula in 1992 23. In 1995, the Guglielmi Detachable Coil (GDC) was approved for use in the US and is now used to treat more than 1100 aneurysms per month worldwide. It is now considered the treatment of choice for most posterior circulation aneurysms. The use of the GDC system for a particular aneurysm is dictated by aneurysm morphology, rather than surgical accessibility. It is generally unsuitable as a stand-alone treatment for large necked or fusiform lesions. However, many investigators have introduced techniques for extending the range of the GDC system. Moret popularized the balloon remodeling technique in which a microcatheter is placed within the aneurysm sac and a microballoon is used to hold the catheter within the aneurysm and protect the vessel lumen from coil herniation 24. Others have used highly navigable coronary stents to reconstruct fusiform arterial lumens and wide-necked aneurysms25-28. Stents provide a barrier for preventing coil herniation into the parent vessel lumen and physically alter blood flow across the aneurysm inflow tract. Within the next few years, prospective randomized trials will compare open surgery to endovascular therapy for the treatment of intracranial aneurysms. Such trials are necessary to better define the role of both surgery and endovascular treatment.

Stroke is the number one cause of adult disability in the industrialized world. As a public health issue, stroke prophylaxis yields tremendous dividends. Carotid endarterectomy (CEA) has been shown by multiple prospective randomized trials to diminish the risk of stroke in carefully selected patients with hemodynamically significant lesions 29. The roles of carotid artery stenting (CAS) and surgical bypass procedures are less clear. Anecdotal case series suggest strongly that CAS may be beneficial in those patients deemed high-risk surgical candidates. Repeat CEA, surgically inaccessible bifurcations, severe coronary artery disease, and radiation induced stenosis are a few of the indications for which CAS may be appropriate 30-32. The role of CAS will be defined in the coming years as it is compared directly and indirectly to surgery in prospective trials. Extracranial-intracranial (EC-IC) bypass was shown to be ineffective by prospective randomized trial in the past33. However EC-IC bypass will be revisited in the coming years as the Carotid Occlusion Surgery Study (COSS) compares best medical therapy to superficial temporal artery to middle cerebral artery bypass in patients with carotid occlusion and documented hemodynamic compromise. Most suspect that EC/IC bypass will be shown to diminish stroke in this high-risk group. Endovascular intracranial angioplasty and stenting for occlusive disease is typically reserved for inoperable or 'high-risk' patients. The past results are variable and anecdotal. Recent series have shown a marked decline in the historically high complication rates and suggest that it may have a role in the future treatment of intracranial atherosclerotic disease34,35.

The role of intravenous tissue plasminogen activator (TPA) has been established for the treatment of acute stroke for patients presenting within 3 h of symptom onset by the National Institute of Neurological Disorders Stroke trial (NINDS)36. In a recent meta-analysis, Brott and colleagues demonstrated greater benefit to those patients receiving intervention within 90 min, reinforcing the widely held perception that time is of the essence. Intra-arterial (IA) therapy has been reserved for those patients presenting outside the intravenous TPA window of 3 h. The efficacy of IA therapy has been proven conclusively in the Prourokinase for Acute Ischemic Stroke Trial37. Acute stroke therapy has been aided by better understanding of the pathophysiology of stroke and the coagulation cascade. Currently, less than 5% of all patients with acute stroke are treated with thrombolytics due to a combination of factors including community education and in-hospital delays. Many institutions capable of delivering i.v. therapy are unable to provide IA therapy to those patients in whom the i.v. therapeutic window has passed. Acute stroke therapy in the future will likely be aided by neuroprotective agents to diminish secondary reperfusion injury as well as thrombolytics and platelet inhibitors delivered by the intravenous and/or intra-arterial route.

Intracranial arteriovenous malformations (AVMs), described as early as 1887 by Pfannenstiel38, remain an often vexing pathology. The first complete excision of an AVM was performed by Pean in a young boy presumed to have a right frontal-parietal tumor 39. Early neurosurgeons advocated extreme conservatism with regard to AVMs. It was not until the introduction of cerebral angiography that surgery became more common as surgeons were able to identify AVMs preoperatively. Pitcher, Olivecrona and Riives, Penfield and Erickson, had all published successful case series by the late 40S 40-42. From 1932 to 1957, nearly 500 patients underwent surgery for AVMs with remarkable success rates. Mortality for small AVMs was approximately 5% and moderate lesions 6%-10%. As complications ensued in patients with incomplete removal of their lesions, the concept of complete removal of AVMs became accepted 43. The first AVM embolization was reported in 1960 by Luessenhop and Spence 44. Several large series of embolizations appeared in the literature reporting varying degrees of success 45,46. In 1977, Stein and Kjellberg et al.43 , presented series of patients treated with radiotherapy. Today, both surgery and radiosurgery play a prominent role in AVM therapy with embolization utilized as adjunctive treatment in specific cases. Endovascular embolization has been greatly aided in North America by the recent FDA approval of NBCA.

The diagnostic state of the art 50 years ago relied on angiographic methods, pneumoencephalography (discovered by Jacobeacus, applied clinically by Dandy), and clinical examination. Hounsfield's introduction of X-ray computed tomography transformed the diagnostic landscape in neurosurgery and neurology. Relative to previous imaging modalities, CT was effectively without complication and allowed brain pathology to be visualized directly rather than by its secondary effects on normal morphology47. Today, we can localize disease and functionality with a combination of noninvasive studies based on the physical and physiologic properties of the central nervous system and its diseased states. While endovascular therapy and cerebral angiography have seen renewed growth, the role of diagnostic cerebral angiography may well diminish as 3-dimensional reformatting of magnetic resonance angiographic and computed tomographic images improves.

In 75 years, neurovascular surgery has progressed in directions which could hardly have been imagined by physicians performing the first angiograms. What the next 75 years will bring is likely to be equally remarkable. The trend towards minimally invasive techniques will undoubtedly continue as budgets for health care strain to cover an increasingly aged population in North America. The ability to minimize morbidity and length of stay in hospital will become ever more important. Some lesions will be deemed best treated by open surgery and others by endovascular methods. This will ultimately be decided by prospective study. We will become better at controlling and minimizing secondary injury during stroke. This may begin simply by applying principles already appreciated by our colleagues caring for those with head injury-- avoidance of hypoxia and control of blood pressure. Arguably, a patient in need of IA thrombolysis should always be under anesthesia for their own benefit in terms of safety, ease of operation, and cerebral protection. Bioactive implants will play an ever increasing role in treatment. Already, our colleagues in cardiology are beginning to utilize coated stents with positive outcomes and soon neuroendovascular surgeons will be able to place bioactive coils within aneurysms to improve neck obliteration. Such treatments are harbingers of the future, encouraging vessels to remodel and repair by controlling local physiologic responses. Just as 75 years ago, some of the future directions of vascular surgery are apparent, but the ultimate destinations are unknown.

ACKNOWLEDGMENTS

Thanks are due to the many authors who have contributed to this issue. The authors are representative of the current state of neurovascular therapy. They are neurologists, neurosurgeons, neurointerventionalists, and basic scientists with often similar interests, complementary and occasionally overlapping skills, whose sole aim is to provide the best possible care to patients. They represent the present and future of our specialties. I must also acknowledge and thank my own mentors and colleagues, Fernando G. Diaz, Manuel Dujovny, and L. Nick Hopkins for their support, wisdom, and encouragement.

REFERENCES

1 Higashida RT, Hopkins LN, Berenstein A, Halbach VV, Kerber C. Program requirements for residency/fellowship education in neuroendovascular surgery/interventional radiology: A special report on graduate medical education. AJNR Am J Neuroradiol 2000; 21: 1153-1159

2 Blane G. History of some cases of disease in the brain, with an account of the appearances upon examination after death, and some general observations on complaints of the head. Transactions: Society for the Improvement of Medical and Chirurgical Knowledge 1800; 2: 192-212

3 Quincke H. Die lumbalpunction in hydrocephalus. Berlin Klinische Wochenschrift 1891; 28: 929-965

4 Moniz E. Aneurysme intra-cranien de la carotide interne droite rendu visible par I'arteriographie cerebrale. Revue Otoneuroophthalmologie 1933; 11: 746-748

5 Moniz E. Die Cerebrale Arterio'raphie and Phlebo raphic, Berlin:

Julius Springer, 1940

6 Cooper A. Account of the first successful operation on the common carotid artery for aneurysm in the year 1808: With the post-mortem examination in 1821. Guy's Hospital Reports 1836; 1: 53-58

7 Zeller 0. Die chirurgische behandlung der durch aneurysma arteriovenosum der carotis int. im sin. cavernosus hervorgerufenen pulsierenden exophthalmos. Deutsch Zeitshrift fur Chirurgie 1911; 111: 1-39

8 Dott N. Intracranial aneurysms: Cerebral arterioradiography: Surgical treatment. Edinburgh Med/ 1933; 40: 219-240 9 Dandy W. Intracranial aneurysm of the internal carotid artery cured by operation. Ann Surg 1938; 107: 654-659

10 Heifetz M. A new intracranial aneurysm clip. J Neurosurg 1969; 30: 753

11 Mayfield FH, Kees G Jr. A brief history of the development of the Mayfield clip. Technical note. J Neurosurg 1971; 35: 97-100

12 Yasargil M. Microsurgery Applied to Neurosurgery, Stuttgart: Georg Thieme, 1969

13 Drake CG, Peerless SJ, Hernesniemi J. Surgery of Vertebrobasilar Aneurysms. London, Ontario experience on 1767 patients, New York: Springer, 1996

14 Velpeau A. Memoire sur la piqure ou I'acupuncture des arteres dans le traitement des aneurismes. Gaz Med Paris 1831; 2: 1-4

15 Phillips B. A Series of Experiments Performed for the Purpose of Showing that Arteries May be Obliterated Without Ligature, Compression or the Knife, London: Longman & Co., 1832

16 Werner SC, Blakemore AH, King BG. Aneurysm of the internal carotid artery within the skull: Wiring and electrothermic coagulation. ]our AMA 1941; 116: 578-582

17 Mullan S, Raimondi AH, Dobben G, Vailati G, Hekmatpanah J. Electrically induced thrombosis in intracranial aneurysms. J Neurosurg 1965; 22: 539-547

18 Luessenhop AJ, Velasquez AC. Observations on the tolerance of the intracranial vessels to catheterization. J Neurosurg 1964; 21: 85-91

19 Serbinenko F. Balloon catheterization and occlusion of major

cerebral vessels. J Neurosurg 1974; 41: 125-145

20 Frei EH, Driller J, Neufeld HN, Barr I, Bleiden L, Askenazy HN. The POD and its applications. Med Res Eng 1966; 5: 11-18

21 Alksne J. Magnetically controlled intravascular catheter. Surgery 1968; 64: 339-345

22 Hilal SK, Michelsen Wj, Driller J, Leonard E. Magnetically guided devices for vascular exploration and treatment. Radiology 1974; 113:529-540

23 Guglielmi G, Vinuela F, Dion J, Duckwiler G. Carotid-cavernous fistula caused by ruptured intracavernous aneurysm: Endovascular treatment by electrothrombosis with detachable coils. Neurosurgery 1992; 31: 591-597

24 Moret J, Cognard C, Weill A, Castaings L, Rey A. Reconstruction technique in the treatment of wide-neck intracranial aneurysms: Long-term angiographic and clinical results -- Apropos of 56 cases. J Neuroradiol 1997; 24: 30-44

25 Higashida RT, Smith W, Gress D, Urwin R, Dowd CF, Balousek PA, Halbach VV. Intravascular stent and endovascular coil placement for a ruptured fusiform aneurysm of the basilar artery: Case report and review of the literature. J Neurosurg 1997; 87: 944-949

26 Lanzino G, Wakhloo AK, Fessler RD, Hartney ML, Guterman LR, Hopkins LN. Efficacy and current limitations of intravascular stents for intracranial internal carotid, vertebral, and basilar artery aneurysms. J Neurosurg 1999; 91: 538-546

27 Mericle RA, Lanzino G, Wakhloo AK, Guterman LR, Hopkins LN. Stenting and secondary coiling of intracranial internal carotid artery aneurysm: Technical case report. Neurosurgery 1998; 43: 1229-1234

28 Sekhon LH, Morgan MK, Sorby W, Grinnell V. Combined

endovascular stent implantation and endosaccular coil placement for the treatment of a wide-necked vertebral artery aneurysm: Technical case report. Neurosurgery 1998; 43: 380-384

29 North American Symptomatic Carotid Endarterectomy Trial Collaborators. Beneficial effect of carotid endarterectomy in symptomatic patients with high-grade stenosis. N Engl J Med 1991; 273: 1421-1428

30 Lopes DK, Mericle RA, Lanzino G, Wakhloo AK, Guterman LR, Hopkins LN. Carotid angioplasty and stenting before coronary artery bypass grafting (Abstract). Neurosurgery 1998; 43: 686

31 Lanzino G, Mericle RA, Lopes DK, Wakhloo AK, Guterman LR, Hopkins LN. Percutaneous transluminal angioplasty and stent placement for recurrent carotid artery stenosis. J Neurosurg 1999; 90:688-794

32 Mericle RA, Kim S, Lanzino G, Lopes D, Wakhloo AK, Guterman

LR, Hopkins LN. Carotid artery angioplasty and use of stents in high-risk patients with contralateral occlusions. J Neurosurg 1999; 90:1031-1036

33 The EC/IC Bypass Study Group. Failure of extracranial-intracranial arterial bypass to reduce the risk of ischemic stroke. Results of an international randomized trial. N Engl J Med 1985; 313: 1191-1200

34 Connors Jj 3rd, Wojak JC. Percutaneous transluminal angioplasty for intracranial atherosclerotic lesions: Evolution of technique and short-term results. J Neurosurg 1999; 91: 415-423

35 Gomez CR, Misra VK, Liu MW, Wadlington VR, Terry JB, Tulyapronchote R, Campbell MS. Elective stenting of symptomatic basilar artery stenosis. Stroke 2000; 31: 95-99

36 National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue plasminogen activator for acute ischemic stroke: The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl IJ Med 1995; 333: 1581-1587

37 Furlan A, Higashida R, Wechsler L, Gent M, Rowley H, Kase C, Pessin M, Ahuja A, Callahan F, Clark WM, Silver F, Rivera F. The PROACT II study - A randomized controlled trial: Prolyse in acute cerebral thromboembolism. JAMA 1999; 282: 2003-2011

38 Pfannenstiel I. Apoplexie als todlicher ausgang von eklampsie. Centralbl Gynaek 1887; 11: 60-61

39 Pean M. Epilepsie partielle symptomatique d'un angiome intracranien des meninges. Trepanation, oblation de la tumeur, guerison. Bull Acad Med (Paris) 1891; 1: 881-883

40 Olivecrona H, Riives J. Arteriovenous aneurysms of the brain. Their diagnosis and treatment. Arch Neurol Psychiat (Chic) 1948; 59: 567-603

41 Penfield W, Ericksen TC. Epilepsy and Cerebral Localization. A Study of the Mechanism, Treatment and Prevention of Epileptic Seizures, Springfield, IL: Thomas, 1941

42 Pitcher C. Angiomatous malformations of the brain. Successful extirpation in three cases. Ann Surg 1946; 123: 239-246

43 Yasargil MG. AVM of the brain, history, embryology, pathological considerations, hemodynamics, diagnostic studies, microsurgical anatomy. In: Yasargil M, ed. Microneurosurgery, lst edn, Vol lilA, Stuttgart: Georg Thieme Verlag, 1987: p. 408

44 Luessenhop AJ, Spence WT. Artificial embolization of cerebral arteries: Report of use in a case of arteriovenous malformation. JAMA 1960; 172: 1153-1155

45 Sano K, Aiba T, Jimbo M. Surgical treatment of cerebral aneurysms and arteriovenous malformations. Neurol med-chir (Tokyo) 1965; 7: 128-131

46 Djindjian R. L'Angiographie Medullaire, Paris: Masson, 1970

47 Webb S. From the Watching Shadows. The Origins of Radiological Tomography, New York: Adam Hilger, IOP Publishing, 1990

Richard D. Fessler, MD Assistant Clinical Professor Director of Neuroendovascular Surgery Harper University Hospital

Departments of Neurosurgery and Radiology Wayne State Univedrsity School of Medicine Detroit, MI, USA

Copyright Forefront Publishing Group 2002
Provided by ProQuest Information and Learning Company. All rights Reserved

Return to Cerebral cavernous malformations
Home Contact Resources Exchange Links ebay