21.03.2013 Views

Craniofacial Anomalies, Part 2 - Plastic Surgery Internal

Craniofacial Anomalies, Part 2 - Plastic Surgery Internal

Craniofacial Anomalies, Part 2 - Plastic Surgery Internal

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

CRANIOFACIAL EMBRYOLOGY<br />

To understand craniofacial anomalies and the context<br />

in which the anomalies occur, a complete<br />

understanding of embryogenesis of this region is<br />

imperative. Sperber1 presents a complete description<br />

of the processes of embryogenesis of the craniofacial<br />

skeleton, excerpted here.<br />

The primary germ layers of the embryo, the ectoderm<br />

and endoderm, form in the inner cell mass of<br />

the blastocyst. The ectoderm differentiates into<br />

cutaneous and neural portions by about day 20.<br />

Neural crest cells make up the most important structures<br />

in craniofacial biology. Neural crest ectomesenchyme<br />

has great migratory propensity and is the major<br />

source of connective tissue throughout the body.<br />

Neural crest cells differentiate into cartilage, bone,<br />

ligaments, muscles, and arteries. Any disruption in<br />

the orderly migration and differentiation of these<br />

cells can have severe consequences, manifested as<br />

congenital defects. 2<br />

The crucial period of organogenesis takes place in<br />

the first 12 weeks of gestation, and it is during this<br />

time that the majority of congenital craniofacial<br />

anomalies occur. 1,2 The earliest signs of the future<br />

face make their appearance at approximately day<br />

23–24 of embryonic life as paired mandibular processes<br />

of the first branchial arch. Neural crest cells<br />

make up the most important structures in the head.<br />

Next, the medial nasal processes combine with the<br />

intervening forebrain to form the frontonasal pro-<br />

CRANIOFACIAL ANOMALIES II:<br />

SYNDROMES AND SURGERY<br />

Delora L Mount MD<br />

cess, which is destined to become the future forehead<br />

and the dorsum of the nose. The lateral nasal<br />

folds separate the olfactory pits from the gradually<br />

developing eye region. By the end of embryonic<br />

week 5, the maxillary and mandibular processes have<br />

begun to increase in size but have not yet fused. It is<br />

not until week 6 that definitive jaws are formed.<br />

By the end of week 8, the face assumes most of<br />

the characteristics that make it recognizable as<br />

human. The face derives from five prominences<br />

that surround the future mouth, the single frontonasal<br />

and the paired maxillary and mandibular processes<br />

(Fig 1).<br />

The grooves between these facial prominences<br />

usually disappear by day 46–47 of gestation. A persisting<br />

groove will result in a congenital facial cleft.<br />

ETIOLOGY OF CRANIOFACIAL ANOMALIES<br />

Table 1 represents suspected contributions to<br />

the development of congenital craniofacial abnormalities.<br />

3,4<br />

The possibility of identifying distinct genetic aberrations<br />

in craniofacial dysmorphology has improved<br />

significantly in recent times. Specific genetic abnormalities<br />

resulting in syndromic craniosynostoses and<br />

facial dysostoses will be presented later in this issue.<br />

In addition to the genetic component, distinct<br />

environmental causes have been identified, includ-<br />

Fig 1. Embryonic development of the human face at (a) 5, (b) 6, and (c) 7 weeks. BG, first branchial groove; FNP, frontonasal prominence;<br />

LNP, lateral nasal prominence; MDP, mandibular prominence; MNP, medial nasal prominence; MXP, maxillary prominence. (Reprinted<br />

with permission from Sperber GH: <strong>Craniofacial</strong> Embryology, 4th ed. London, Wright, 1989.)


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

ing radiation, infection, maternal factors, and chemical<br />

exposures.<br />

Radiation. Large doses of radiation have been<br />

associated with microcephaly.<br />

Infection. The children of mothers affected with<br />

toxoplasmosis, rubella, or cytomegalovirus show<br />

increased frequency of facial clefts as well as concomitant<br />

hand and ocular abnormalities.<br />

Maternal idiosyncrasies. Mothers of cleft lip and<br />

palate children have been noted to have a higherthan-normal<br />

incidence of the phenylketonuria disorder.<br />

The oculoauriculovertebral (OAV) spectrum has<br />

been seen with unusual frequency in infants of diabetic<br />

mothers. 5 Many studies associate maternal<br />

factors such as age, weight, and general health as<br />

potential causes of malformation.<br />

Chemicals. Vitamin deficiency states are associated<br />

with an increased incidence of cleft lip and<br />

2<br />

TABLE 1<br />

Causes of Congenital Deformities in Man<br />

(Reprinted with permission from Slavkin HC: Developmental<br />

<strong>Craniofacial</strong> Biology. Philadelphia, Lea & Febiger, 1979.)<br />

palate; which may be reduced with vitaminsupplementation<br />

diets for the mothers. Numerous<br />

studies have demonstrated a reduced incidence of<br />

facial clefts after maternal supplementation with folic<br />

acid. 6<br />

Vitamin A, its derivatives, and related compounds<br />

such as isotretinoin (Accutane) have been implicated<br />

in the development of facial clefting and hemifacial<br />

microsomia. Maternal smoking is associated with<br />

craniosynostosis 7 and facial clefts. 8<br />

Additional substances are implicated in increased<br />

risk of craniofacial anomalies, such as chlorpheniramine,<br />

chlordiazepoxide, and nitrofurantoin exposure<br />

and craniosynostosis. 9<br />

CLASSIFICATION<br />

To date no single classification system has been<br />

devised that accurately describes all congenital craniofacial<br />

anomalies. In 1981 the Committee on<br />

Nomenclature and Classification of <strong>Craniofacial</strong><br />

<strong>Anomalies</strong> of the American Cleft Palate Association10 grouped craniofacial disorders according to their<br />

diverse etiology, anatomy, and treatment. They propose<br />

a practical and simple classification system in<br />

which five categories of deformity are identified, as<br />

follows:<br />

I Clefts (Tessier classification)<br />

II Synostoses<br />

III Atrophy/hypoplasia<br />

IV Neoplasia/hyperplasia<br />

V Unclassified<br />

I — CLEFTS, DYSPLASIAS, AND DYSOSTOSES<br />

Anatomic Classification<br />

In 1976 Tessier11 described an anatomic classification<br />

system whereby a number is assigned to each<br />

of the malformations according to its position relative<br />

to the sagittal midline (Fig 2).<br />

For orientation, the orbit is divided into two hemispheres:<br />

The lower lid with cheek and lip demonstrate<br />

facial clefts, the upper lid demonstrates cranial<br />

clefts. Two Tessier classification schemes exist, one<br />

for the skeleton and one for soft tissue. Numerous<br />

instances may occur where there is discrepancy<br />

between the two classification systems—for example,<br />

a subject with a bony cleft but no soft-tissue cleft, or


CENTRIC<br />

Corresponding Cranial<br />

Facial Clefts Extension of Facial Clefts<br />

No. 0 No. 14<br />

No. 1 No. 13<br />

No. 2 No. 12<br />

No. 3 No. 11<br />

ACENTRIC<br />

Corresponding Cranial<br />

Facial Clefts Extension of Facial Clefts<br />

No. 4 No. 10<br />

No. 5 No. 9<br />

No. 6<br />

No. 7<br />

No. 8<br />

Fig 2. Tessier’s classification of craniofacial clefts. Localization on<br />

the soft tissues (above) and skeleton (below). (Reprinted with<br />

permission from Tessier P: Anatomical classification of facial,<br />

cranio-facial, and latero-facial clefts. J Maxillofac Surg 4:69,<br />

1976; list from Whitaker LA, Pashayan H, and Reichman J: A<br />

proposed new classification of craniofacial anomalies. Cleft<br />

Palate J 18:161, 1981.)<br />

where skeletal cleft and soft-tissue cleft are not in the<br />

same position. Nevertheless, Tessier’s scheme remains<br />

in wide use today because it is relatively easy to learn<br />

for communicating with other clinicians. David et<br />

al 12 illustrate a complete series of these craniofacial<br />

clefts in 3D CT scans.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

The tissue-deficiency disorders—arhinencephaly<br />

and holoprosencephaly—are secondary to failure of<br />

cleavage of the embryonic holoprosencephalon and<br />

of the normal longitudinal split into cerebral hemispheres.<br />

The tissue-excess deformities range from a<br />

slight midline notch of the upper lip to severe orbital<br />

hypertelorism.<br />

The holoprosencephaly malformation represents<br />

a hypoplastic No. 14 cleft in association with a tissue<br />

deficiency or a tissue excess. 13,14 Cohen and Sulik 15<br />

present a modern analytic review of the holoprosencephalic<br />

disorders. Central nervous system<br />

findings and craniofacial anatomy are discussed, syndromes<br />

and associated anomalies are updated, and<br />

the differential diagnosis is reviewed.<br />

Elias, Kawamoto, and Wilson 16 reviewed holoprosencephaly<br />

and midline facial anomalies in an<br />

attempt to redefine their classification and management.<br />

They note that true holoprosencephaly<br />

encompasses a series of midline defects of the brain<br />

and face, and in most cases is associated with severe<br />

malformations of the brain which are incompatible<br />

with life. At the other end of the spectrum are<br />

patients with midline facial defects and normal or<br />

near-normal brain development.<br />

Embryologic Classification<br />

Van der Meulen and coworkers13 tried to correlate<br />

clinical features of the disorders with embryologic<br />

events.<br />

Site of dysplasia/dysostosis and associated cleft:<br />

Frontosphenoidal = Tessier 9<br />

Frontal dysplasia = Tessier 10 and 11<br />

Interfrontal dysplasia = Tessier 0 and 14<br />

Treacher Collins = 6, 7, and 8 clefts<br />

Temporoauromandibular dysplasia = Hemifacial<br />

microsomia (craniofacial microsomia)<br />

Pathogenesis of Clefts<br />

There are two leading theories of facial cleft formation.<br />

The classic theory holds that clefts are caused<br />

by failure of fusion of the facial processes. 17-19 In this<br />

theory the medial face unites by fusion of the paired<br />

facial processes beneath the nasal pits. Epithelial<br />

contact is established and mesenchymal penetration<br />

completes the fusion of the lip and palate. If the<br />

sequence is disturbed, a cleft forms.<br />

3


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

The mesodermal penetration theory states that<br />

the edges of the facial processes consist of a bilaminar<br />

membrane of ectoderm with epithelial seams<br />

demarcating the major process. 20-23 Mesenchymal<br />

cells then penetrate the layers and smooth out the<br />

seams. If the mesenchymal penetration fails, the<br />

epithelium dehisces and a cleft results. The severity<br />

of the cleft is proportional to the amount of mesodermal<br />

penetration.<br />

Management of Facial Clefts<br />

By far the most common craniofacial anomaly is<br />

cleft of the lip/palate, followed by isolated cleft palate<br />

as a distant second. These topics have been<br />

discussed in detail in Selected Readings in <strong>Plastic</strong><br />

<strong>Surgery</strong> volume 10, number 16, 24 and will not be<br />

addressed further here.<br />

The incidence of rare clefts is estimated at 1.4–4.9<br />

per 100,000 live births. 25 Reconstruction focuses<br />

initially on soft-tissue closure25 with excision of the<br />

free borders of the cleft to normal tissue, followed by<br />

meticulous layered soft-tissue closure.<br />

Van der Meulen26 offers a thorough review of the<br />

pathology, etiology, and reconstruction of oblique<br />

facial clefts. He agrees with Tessier’s principle of<br />

combining skeletal and soft-tissue realignment in one<br />

major surgical procedure. Resnick and Kawamoto, 27<br />

Galante and Dado, 28 and Fuente del Campo29 give<br />

specific recommendations for the treatment of<br />

unusual facial clefts on the basis of their respective<br />

experiences with Tessier’s Nos. 4, 5, and 8 clefts.<br />

Menard30 describes the application of tissue<br />

expansion in the soft-tissue closure of facial clefts in 8<br />

patients. Due to underlying bony hypoplasia, skeletal<br />

reconstruction is often necessary when the child<br />

is older. 25<br />

There are multiple approaches to cleft repair, timing<br />

of repair, and technique of reconstruction.<br />

Although several protocols have been suggested, these<br />

should be viewed as guidelines, not absolute directives<br />

for care. The variability in approaches mirrors<br />

the extreme variability in congenital clefts, dysplasias,<br />

and dysostoses. A few in-depth details and reconstructive<br />

algorithms will be presented for the more<br />

common abnormalities, including craniofacial<br />

microsomia, Goldenhar syndrome, Treacher Collins<br />

syndrome, Nager syndrome, Binder syndrome, Pierre<br />

Robin sequence, and encephaloceles.<br />

4<br />

CRANIOFACIAL MICROSOMIA<br />

<strong>Craniofacial</strong> or hemifacial microsomia is the term<br />

most frequently used to describe the first and second<br />

branchial arch syndrome, which is defined as a Tessier<br />

7 atypical facial cleft. Thomson31 was the first to<br />

suggest in 1843 that the malformation was due to<br />

imperfect development of the first two anterior branchial<br />

arches. Clinical manifestations are underdevelopment<br />

of the external and middle ear, mandible,<br />

zygoma, maxilla, temporal bone, facial muscles,<br />

muscles of mastication, palatal muscles, tongue, and<br />

parotid gland; macrostomia; a first branchial cleft<br />

sinus; 31–33 and possible involvement of any or all<br />

cranial nerves34,35 (Fig 3).<br />

Fig 3. <strong>Craniofacial</strong> microsomia in a 7-year-old child. (Reprinted<br />

with permission from McCarthy JG, Grayson BH: Reconstruction:<br />

<strong>Craniofacial</strong> Microsomia. In Mathis SJ (ed), <strong>Plastic</strong><br />

<strong>Surgery</strong>, 2nd ed. Philadelphia, Elsevier, 2006. Vol IV, Ch 103,<br />

pp 521-554.)<br />

The birth incidence of craniofacial microsomia is<br />

approximately 1 in 4000. 35 Approximately 10% of<br />

cases are bilateral. 31,36 The etiology is thought to<br />

relate to occlusion or thrombosis of the stapedial<br />

artery with injury to the developing first and second<br />

branchial arches. 35,37 In its fullest expression, the<br />

craniofacial microsomia syndrome is made up of a<br />

constellation of congenitally malformed facial structures<br />

arising from the embryonic first and second


visceral arches, the intervening first pharyngeal pouch<br />

and first branchial cleft, and the primordia of the<br />

temporal bone. 38<br />

Originally craniofacial microsomia was thought to<br />

represent a progressive skeletal and soft-tissue deformity<br />

that worsens over time, 39 but subsequently<br />

Polley 40 assessed longitudinal cephalometric data from<br />

26 patients with unoperated hemifacial microsomia<br />

and demonstrated that the condition is not<br />

progressive. These findings were further confirmed<br />

by Kearns et al 41 in 67 subjects. The disorder varies<br />

widely in presentation and may range from simple<br />

preauricular skin tags to composite mandibular and<br />

maxillary hypoplasia. Its management depends on<br />

the severity of the defect and the functional and<br />

aesthetic reconstructive needs. 42-46<br />

Pruzansky 44 described three types of mandibular<br />

deficiency in craniofacial microsomia according to<br />

the anatomical area affected (Table 2).<br />

This classification was modified by Mulliken and<br />

Kaban, 47 who subdivided Type II into Type IIA, in<br />

which the glenoid fossa-condyle relationship is maintained<br />

and the TMJ is functional, and Type IIB, in<br />

which the glenoid fossa-condyle relationship is not<br />

maintained and the TMJ is nonfunctional.<br />

Munro’s 42,44 classification extended the skeletal<br />

anomaly to include the orbit. This system aims at<br />

providing a basis for surgical reconstruction and consists<br />

of five types denoting increasingly severe hypoplasia<br />

of the facial bones (Table 3). Isolated microtia<br />

is considered to be a microform of craniofacial<br />

microsomia. 48<br />

Meurman 46 recognizes three grades of auricular<br />

deformity, as follows: Grade I: distinctly smaller,<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

malformed auricle but all components are present;<br />

Grade II: only a vertical remnant of cartilage and<br />

skin is present, with atresia of the external meatus;<br />

Grade III: complete or nearly complete absence of<br />

the auricle.<br />

David and colleagues 49 proposed a multisystem<br />

classification of hemifacial microsomia in the TNM<br />

style. The physical manifestations of hemifacial<br />

microsomia are graded according to five levels of<br />

skeletal deformity (S1–S5) equivalent to the Pruzansky<br />

classification for S1-S3, plus S4 representing orbital<br />

involvement and S5 representing orbital dystopia.<br />

Auricular deformity (A0–A3) is similar to the classification<br />

described by Meurman. Tissue deficiency<br />

(T1–T3) is graded as mild, moderate, or severe. The<br />

SAT sytem allows a comprehensive and staged<br />

approach to skeletal and soft-tissue reconstruction.<br />

Early macrostomia repair (1–2 months of age)<br />

yields excellent functional and cosmetic results. 50<br />

More extensive reconstruction, including composite<br />

correction in moderate to severe deformity, is reserved<br />

for early childhood (age 5–6) but should not wait<br />

until facial growth is complete. 39,42–44,51–54 The mandible<br />

is usually corrected first in the hope that repositioning<br />

the jaw will unlock the growth potential of<br />

the functional matrix to allow normal growth of the<br />

mandible 55,56 and release abnormal growth tendencies<br />

of the maxilla.<br />

In mild cases, Posnick prefers to wait for skeletal<br />

maturity, and employs traditional orthognathic<br />

surgery to achieve favorable aesthetic results. 50 Distraction<br />

osteogenesis provides excellent correction<br />

in cases of mandibular deformity up to Type IIB. 57,58<br />

In cases of Type III deformity, a costochondral<br />

TABLE 2<br />

Mandibular Deficiency in <strong>Craniofacial</strong> Microsomia (Pruzansky)<br />

5


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

graft is usually indicated, though many authors have<br />

noted unpredictable overgrowth of costochondral<br />

grafts. 34,42–44,53,59<br />

Ross 60 reviewed 55 cases of severe craniofacial<br />

microsomia treated with costochondral grafts at<br />

The Hospital for Sick Children. The success rates<br />

were higher when the children were operated earlier<br />

(85% for age 3–7y vs 50% for age >14y).<br />

Growth equal to the other side was seen in 46%,<br />

undergrowth in 15%, and overgrowth in 39%.<br />

Given the option of early or delayed surgery, the<br />

author favors early surgery at age 4–5, citing a<br />

higher graft success rate, the psychosocial advantage<br />

of attaining facial symmetry at a younger age,<br />

and the additional benefit that erupting teeth will<br />

assume a more normal position, making future<br />

orthodontic treatment less difficult.<br />

Munro 42,44,53 usually advocates much more<br />

extensive surgery, operating on the maxilla at the<br />

same time as the mandible. For mild cases, mandibular<br />

surgery may involve contour surgery with or<br />

without genioplasty once skeletal maturity is<br />

reached. In case of orbitozygomatic hypoplasia,<br />

Posnick 50 uses split cranial bone grafts at age 5–7,<br />

stating that at age 7 the cranioorbitozygomatic complex<br />

is nearly mature so that an adult-size vault,<br />

orbit, and cheekbone can be fashioned. Also the<br />

thickness of the calvarium at that age makes for an<br />

easier harvest of split calvarial bone grafts. Refinements<br />

to the mature skeleton may be needed eventually<br />

to achieve a symmetrical and aesthetic<br />

result, 50 and may take the form of sagittal split<br />

osteotomy, Le Fort I osteotomy, or genioplasty.<br />

At times the soft-tissue deformity in craniofacial<br />

microsomia must also be addressed, and usually follows<br />

skeletal reconstruction. Various methods of<br />

6<br />

TABLE 3<br />

Mandibular and Orbital Deficiency in <strong>Craniofacial</strong> Microsomia (Munro)<br />

soft-tissue augmentation with microvascular free transfers<br />

are described by La Rossa 61 and Upton. 62 This<br />

subject is further discussed in Selected Readings in<br />

<strong>Plastic</strong> <strong>Surgery</strong> volume 10, number 5, part 1. 63<br />

GOLDENHAR SYNDROME<br />

(Oculoauriculovertebral Dysplasia)<br />

Goldenhar syndrome is characteristically bilateral.<br />

Features include prominent frontal bossing,<br />

a low hairline, mandibular hypoplasia, low-set ears,<br />

colobomas of the upper eyelid, epibulbar dermoids,<br />

accessory auricular appendages that are bilateral<br />

and anterior to the ears, and vertebral anomalies. 5<br />

Occurrence is believed to be sporadic, with only a<br />

weak genetic component. The presence of<br />

epibulbar dermoid is required for a diagnosis of<br />

Goldenhar syndrome. The reconstruction follows<br />

the principles of craniofacial microsomia presented<br />

above.<br />

TREACHER COLLINS SYNDROME<br />

(Mandibulofacial Dysostosis)<br />

The first reference to mandibulofacial dysostosis<br />

in the medical literature was made by Berry in 1889.<br />

Berry described the physical symptoms and speculated<br />

about the inherited character of the deformity.<br />

His treatise was certainly much more detailed than<br />

the two cases reported by E Treacher Collins 11<br />

years later, after whom the syndrome was named.<br />

In Europe the deformity is known as the Franceschetti-<br />

Zwahlen-Klein syndrome on the basis of their 1949<br />

monograph summarizing the world literature. 64 The<br />

incomplete form should be designated as Treacher<br />

Collins syndrome65 and the complete form as<br />

Franceschetti’s syndrome.


The Treacher Collins syndrome66 represents a<br />

manifestation of the Tessier Nos. 6, 7, and 8 cleft. It<br />

is inherited as an autosomal dominant trait with an<br />

incidence of 1/10000 live births. 67 Bilaterality is the<br />

norm and phenotypic expression is variable. The<br />

gene for Treacher Collins syndrome was identified<br />

in 1991, when it was mapped to chromosome 5 by<br />

Dixon and colleagues68 from a study of 12 families.<br />

Later that year the genetic locus was refined to bands<br />

5q31.3?q33.3. 69 In 1996, transcription mapping<br />

localized the critical region to a specific locus, TCOF1,<br />

which produces an abnormal protein named Treacle.<br />

This nucleolar protein is under intense study and<br />

appears to function in microtubule formation and<br />

cell migration. 70,71<br />

The pathogenesis of Treacher Collins syndrome<br />

remains unknown. Sulik and colleagues72 were able<br />

to produce the facial abnormalities of Treacher Collins<br />

in mice by administering isotretinoin, suggesting that<br />

the syndrome can be triggered by disruption of vitamin<br />

A metabolism. There is an apparent correlation<br />

between frequency of mutation and advanced<br />

paternal age.<br />

The typical features of the Treacher Collins syndrome<br />

include the following (Fig 4):<br />

• palpebral fissures sloping downward laterally<br />

(antimongoloid slant), with coloboma of the outer<br />

portion of the lower lid and rarely the upper lid<br />

• hypoplasia (aplasia) of the facial bones, especially<br />

the malar bones and mandible<br />

• malformation of the external ear and occasionally<br />

the middle and inner ear<br />

Fig 4. Treacher-Collins Syndrome: Softtissue<br />

and skeletal deformities. (Reprinted<br />

with permission from Bartlett SP, Losee JE,<br />

Baker SB: Reconstruction: <strong>Craniofacial</strong> Syndromes.<br />

In Mathis SJ (ed), <strong>Plastic</strong> <strong>Surgery</strong>,<br />

2nd ed. Philadelphia, Elsevier, 2006. Vol IV,<br />

Ch 102, pp 495-519.)<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

• macrostomia, high palate, abnormal position and<br />

malocclusion of the teeth<br />

• blind fistula between the angles of the mouth<br />

and the ears<br />

• atypical hair growth in the form of tongue-shaped<br />

processes of the hairline extending toward the<br />

cheeks<br />

• absence of eyelashes in at least the medial third<br />

of the lower eyelid<br />

In affected newborns the first priority is airway<br />

management. Shprintzen et al 73 noted that some<br />

patients have marked narrowing of the airway (pharyngeal<br />

diameter


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

traction osteogenesis in children


Fig 5. (Above) The nose and upper lip in maxillonasal dysplasia.<br />

1 - Retracted columella-lip junction and lack of triangular flare at<br />

the base. 2 - Perpendicular alar-cheek junction. 3 - Upper lip<br />

convex with wide, shallow philtrum. 4 - Crescent-shaped nostril<br />

without nostril sill. 5 - Low set and flat nasal tip. 6 - Cupid’s bow<br />

stretched and shallow. (Below) Surgical correction is by medial<br />

rotation of tissues on either side of the nasal midline. (Reprinted<br />

with permission from Holmstrom H: Clinical and pathologic<br />

features of maxillonasal dysplasia (Binder’s syndrome): significance<br />

of the prenasal fossa on etiology. Plast Reconstr Surg<br />

78:559, 1986.)<br />

a coronal incision and reaching the nasal floor through<br />

an incision in the upper buccal sulcus. The nasal soft<br />

tissues and alar cartilages are mobilized. The nose is<br />

lengthened and tip projection is achieved with a cantilever<br />

graft of lyophilized cartilage.<br />

Wolfe 107 describes a technique of nasofrontal<br />

osteotomy to lengthen the nose in cases of posttraumatic<br />

shortening and Binder syndrome.<br />

McCollum 108 reviews the literature and provides long<br />

term follow up of 2 patients, one treated with traditional<br />

orthognathic surgery and the other with a<br />

growth center implant to the nose.<br />

PIERRE ROBIN SEQUENCE<br />

In 1923 Pierre Robin, a French stomatologist, noted<br />

a triad of characteristics of the upper airway which is<br />

now known as the Pierre Robin sequence. 109 The<br />

characteristic features consist of micrognathia,<br />

glossoptosis, and airway obstruction. 8,109 An associated<br />

high arched midline cleft of the soft palate and occasionally<br />

of the hard palate is present in approximately<br />

50% of cases. 110,111 The sequence shows great etiologic<br />

heterogeneity, with as many as 18 associated syndromes.<br />

The glossoptosis in Pierre Robin can begin a vicious<br />

sequence of events: airway obstruction, increased<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 6. Patient with Binder syndrome. A, B, at age 10. C, E,<br />

preoperatively at age 17. D, F, after orthodontic treatment (maxillary<br />

first bicuspid extractions), orthognathic surgery (Le Fort I osteotomy<br />

with horizontal advancement), and nasal reconstruction<br />

(corticocancellous iliac graft). (Reprinted with permission from<br />

Posnick JC, Tompson B: Binder syndrome: staging of reconstruction<br />

and skeletal stability and relapse patterns after Le Fort I osteotomy<br />

using miniplate fixation. Plast Reconstr Surg 99:967, 1997.)<br />

energy expenditure, and decreased caloric intake<br />

from impaired feeding. Afflicted infants typically fail<br />

to thrive because of respiratory and feeding difficulties.<br />

If these problems are ignored, respiratory failure,<br />

cardiac failure, and death may result.<br />

9


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

In 1946 Douglas 112 reported >50% mortality with<br />

conservative treatment of Pierre Robin. It is now<br />

clear that the key to successful medical treatment of<br />

infants with Pierre Robin is to hold the infant prone<br />

to relieve the glossoptosis and open the airway. In<br />

some cases this position must be maintained for 24<br />

hours a day, even during feeding, baths, and diaper<br />

changing. 113<br />

While most infants can be successfully managed<br />

conservatively, a few will require surgical intervention.<br />

If medical treatment fails to relieve the symptoms<br />

of airway obstruction, the baby would formerly<br />

be considered for tongue-lip adhesion or tracheostomy.<br />

112,114,115 The advent of distraction osteogenesis<br />

provides an effective alternative for addressing<br />

airway obstruction. Denny 116 describes a series of<br />

10 patients with airway obstruction from severe craniofacial<br />

syndromes who were treated with distraction<br />

osteogenesis of the mandible. All children were clinically<br />

improved, and 2 of 3 patients with tracheostomies<br />

were successfully decannulated within 6 weeks.<br />

The mean functional airway increase after distraction<br />

was 67.5% (Fig 7).<br />

ENCEPHALOCELES<br />

An encephalocele is a protrusion of part of the<br />

cranial contents through a defect in the skull. The<br />

mass may contain meninges (meningocele), meninges<br />

and brain (meningoencephalocele), or meninges,<br />

brain, and ventricle (meningoencephalocystocele). 117<br />

Encephaloceles categorized by their position in the<br />

skull can be basal, sincipital, or convexity. 118 The<br />

sincipital group can be further divided into<br />

frontoethmoidal, interfrontal, and those associated<br />

with clefts. The frontoethmoidal group can be subdivided<br />

into nasofrontal, nosoethmoidal and<br />

nasoorbital types. 119<br />

The presence of an encephalocele may be detected<br />

on fetal ultrasound or by an elevated alpha fetoprotein<br />

level. 118 The differential diagnosis of frontal midline<br />

masses includes encephaloceles, teratomas, gliomas,<br />

and dermoids. 120,121 High-resolution CT scans<br />

can establish the intracranial component of<br />

encephaloceles. 121 In a frontoethmoidal or nasal<br />

encephalocele, the cranial defect is in the anterior<br />

midline between the frontal bone preformed in membrane<br />

and the ethmoid preformed in cartilage. The<br />

craniofacial deformity consists of hypertelorism, orbital<br />

dystopia, elongation of the face, and dental maloc-<br />

10<br />

Fig 7. Two patients with Nager syndrome and tracheostomy before<br />

(left) and after (right) mandibular distraction. (Reprinted with<br />

permission from Denny AD, Talisman R, Hanson PR, Recinos RF:<br />

Mandibular distraction osteogenesis in very young patients to<br />

correct airway obstruction. Plast Reconstr Surg 108:302, 2001.)<br />

clusion, reflecting the distorting influences on facial<br />

bone growth by the extruded intracranial contents<br />

(Fig 8).<br />

The pathogenesis of frontoethmoidoencephaloceles<br />

is as follows. Early in embryogenesis, diverticula<br />

of dura project anteriorly through the fonticulus<br />

nasofrontalis (a small fontanelle between the developing<br />

nasal and frontal bones) or inferiorly through<br />

the developing frontal bone into the prenasal space.<br />

These diverticula may come in contact with skin and<br />

adhere to it. Normally the diverticulum regresses<br />

and the bone closes, creating both the normal<br />

nasofrontal suture anteriorly and, passing through<br />

the skull base just anteriorly to the crista galli, the


Fig 8. Left, child with asymmetrical nasoethmoidal, nasoorbital<br />

encephalocele. Right, same child at age 5, after removal of the<br />

encephalocele and correction of the trigonocephaly, orbital<br />

dystopia, and hypertelorism in one procedure at 8 months of age.<br />

(Reprinted with permission from Holmes AD, Meara JG, Kolker<br />

AR, et al: Frontoethmoidal encephaloceles: reconstruction and<br />

refinements. J Craniofac Surg 12:6, 2001.)<br />

foramen cecum. In a frontoethmoidal encephalocele<br />

the diverticulum does not recede and the bone<br />

does not close. The etiology of encephalocele is<br />

unknown but includes racial, genetic, environmental,<br />

and paternal factors. 122<br />

Encephaloceles occur with a number of craniofacial<br />

syndromes. 123 The world wide incidence of<br />

encephalocele is 1/5000. 124 In Western Europe, North<br />

America, Australia, and Japan, occipital encephaloceles<br />

predominate. In Southeast Asia and Russia,<br />

anterior encephaloceles outnumber posterior ones<br />

by a 9.5:1 ratio. 124,125 The reason for this discrepancy<br />

is unknown.<br />

The principles of treatment are incision of the sac,<br />

amputation of excess tissue to the level of the surrounding<br />

skull, closure of the dura, and closure of<br />

the skin. 126 David, 127 Forcada et al, 128 and Smit and<br />

colleagues129 review the spectrum of cranial and<br />

cerebral malformations that may be present in<br />

frontoethmoidal encephaloceles and discuss the<br />

diagnosis and management of these deformities.<br />

David127 analyzed the experience with frontoethmoidal<br />

encephaloceles by the Australian <strong>Craniofacial</strong><br />

Unit from 1975 to 1993 and reached the following<br />

conclusions:<br />

• Early complete surgery is indicated to allow the<br />

developing brain and eyes to remodel the facial<br />

deformity.<br />

• Intracranial abnormalities are common.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

• Frontoethmoidal encephaloceles differ from other<br />

neural tube defects in their lack of a familial pattern<br />

and peculiar geographic distribution.<br />

• Treatment by craniofacial technique is best.<br />

• The established deformity can be effectively managed<br />

by craniofacial osteotomies.<br />

• Most patients have abnormal intercanthal distances<br />

but normal interpupillary and lateral canthal<br />

measurements.<br />

• The frontal sinus region often needs repeat bone<br />

grafting, and nasal bone grafts commonly need<br />

to be replaced as patients age.<br />

• Treatment for craniofacial clefts should be postponed<br />

until after growth is complete.<br />

• Early treatment of patients with basal encephaloceles<br />

is indicated to prevent further damage and<br />

infection.<br />

• <strong>Surgery</strong> for extensive basal encephaloceles is complex<br />

and probably should be done through a<br />

facial hemisection approach.<br />

Holmes et al130 offer their experience with 35<br />

cases of frontoethmoidal encephalocele. The goals<br />

of treatment are as follows (Fig 9):<br />

• urgent closure of open skin defects to prevent<br />

infection and desiccation of brain<br />

• removal or invagination of nonfunctional extracranial<br />

tissue<br />

• watertight dural closure<br />

• total craniofacial reconstruction with special care<br />

to avoid the “long nose deformity”<br />

To correct the deformity caused by hypertelorism<br />

and a long midface, Holmes and coworkers130 lower<br />

the supraorbital bar by rotating it medially, posteriorly<br />

and downward in the midline, while laterally it is<br />

widened to correct the trigonocephalic deformity.<br />

Successful correction depends on an understanding<br />

of the pathologic anatomy; careful planning of<br />

osteotomies and bone movements to correct the<br />

whole deformity, including trigonocephaly and the<br />

long nose deformity; nasal reconstruction with cantilever<br />

graft to avoid the long nose deformity; skin<br />

closure removing abnormal skin and careful placing<br />

of scars; transnasal canthoplasty to reposition the<br />

11


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 9. Three-dimensional models of a frontoethmoidal encephalocele and its correction. A, B, note the external cranial opening, interorbital<br />

hypertelorism, and depressed cribriform plate. The supraorbital osteotomies and central metopic area of bone are marked, as well as the<br />

bony Z-plasties on the lateral orbital rims. C, diagram of the proposed osteotomies and bone movements. D, after repositioning the medial<br />

and lateral walls of the orbit and supraorbital bar and fixation of the bone graft for nasal reconstruction. (Reprinted with permission from<br />

Holmes AD, Meara JG, Kolker AR, et al: Frontoethmoidal encephaloceles: reconstruction and refinements. J Craniofac Surg 12:6, 2001.)<br />

medial canthi; and single-stage surgery displaying craniofacial<br />

and neurosurgical expertise.<br />

II — CRANIOSYNOSTOSIS<br />

Virchow (1851) was the first to use the term craniosynostosis,<br />

although abnormal head shapes related<br />

to cranial sutures were noted by Hippocrates as early<br />

as 100 BC. Virchow noted that growth restriction<br />

occurred perpendicular to the fused cranial suture<br />

and compensatory growth occurred parallel to the<br />

affected suture 131 (Fig 10). This combination of growth<br />

restriction in one dimension and compensatory<br />

growth at right angles results in consistent patterns of<br />

abnormal head shape. 132<br />

12<br />

The incidence of craniosynostosis is estimated at 1<br />

in 2000 live births. 133 Retrospective studies using<br />

radiographic criteria alone underestimate the occurrence<br />

of craniosynostosis. 134,135 Craniosynostosis may<br />

occur as a sporadic, isolated abnormality or as a feature<br />

of a congenital syndrome. It can be isolated<br />

nonsyndromic or syndromal. The best way to classify<br />

isolated craniosynostosis is to name the suture(s)<br />

involved—eg, left unicoronal synostosis, metopic synostosis,<br />

bicoronal synostosis—but it is also common to<br />

refer to the resulting head shape 123,136–138 (Fig 11).<br />

Trigonocephaly results from premature fusion of<br />

the metopic suture. The spectrum of deformities<br />

includes a vertical midline forehead ridge, triangular<br />

or “keel-shaped” contour, bitemporal narrowing, and<br />

hypotelorism.


Fig 10. Cranial sutures in the human fetus. Premature closure<br />

produces growth restriction perpendicular to the line of the<br />

suture and compensatory overgrowth parallel to it.<br />

Scaphocephaly (dolichocephaly) is due to premature<br />

fusion of the sagittal suture. Features of sagittal<br />

synostosis include a palpable ridge overlying the sagittal<br />

suture, decreased biparietal diameter, and elongation<br />

of the skull in the anteroposterior dimension.<br />

Significant frontal and occipital bossing are commonly<br />

noted. The appearance resembles a boat or “scaphe.”<br />

Plagiocephaly stems from the Greek word meaning<br />

“crooked head”, and is an asymmetrical deformity.<br />

It may be anterior or posterior. Two etiological<br />

variants must be distinguished – deformational<br />

and synostotic.<br />

Posterior plagiocephaly is usually positional and a<br />

result of the child lying predominately on his/her<br />

back. It produces a classic parallelogram skull<br />

deformity. 139 The incidence has increased with the<br />

recommendation by the American Academy of<br />

Pediatrics to lay infants on their backs to reduce the<br />

risk of sudden infant death syndrome (SIDS). 140,141<br />

Treatment of positional plagiocephaly depends on<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

the severity of the deformity and often requires helmet<br />

therapy for correction in severe cases. However,<br />

large studies have shown little difference in<br />

outcome between helmet therapy and consistent<br />

repositioning of the infant off the flat spot in mild to<br />

moderately severe cases. 141,142<br />

Posterior synostotic plagiocephaly can also result<br />

from unilambdoid synostosis. Unilambdoid synostosis<br />

is a very rare entity.<br />

Brachycephaly is the result of bicoronal synostosis<br />

and is characterized by anteroposterior shortening<br />

of the skull. The lower part of the forehead and<br />

supraorbital bar are retropositioned. This is the<br />

characteristic head shape deformity that accompanies<br />

Apert and Crouzon syndrome, and in these<br />

cases it is believed to be due to abnormalities that<br />

extend into the cranial base, causing the associated<br />

facial deformities of exorbitism and maxillary retrusion.<br />

Posterior brachycephaly is unusual but can<br />

be the external manifestation of bilateral lambdoid<br />

suture synostosis.<br />

Turricephaly (towering head deformity) is characterized<br />

by excessive skull height and a vertical forehead.<br />

This deformity is typically an untreated<br />

brachycephaly where compensatory expansion leads<br />

to an increasing vertical height to the cranium. Children<br />

with Apert syndrome have a particular tendency<br />

toward turricephaly.<br />

Oxycephaly is a pointed head. The forehead is<br />

retroverted and tilted back in continuity with the<br />

nasal dorsum. Oxycephaly is usually due to fusion of<br />

multiple sutures.<br />

When multiple sutures are involved, head shapes<br />

are more variable and the distinctions blur. In these<br />

cases the specific sutures involved should be named<br />

when describing the deformity. The kleeblattschadel<br />

or cloverleaf skull deformity results from pansutural<br />

synostosis, and usually requires early, aggressive treatment<br />

to prevent cerebral compromise.<br />

<strong>Craniofacial</strong> Growth<br />

The cranium is made up of the neurocranium,<br />

which includes the chondrocranium of the skull base<br />

and the membranous bone of the calvarium, and<br />

the viscerocranium, which forms the membranous<br />

bones of the face. The various areas of the<br />

craniomaxillofacial skeleton grow by very different<br />

methods. The cranial sutures are skeletal joints of<br />

the syndesmosis type as well as being sites of osteo-<br />

13


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 11. Skull shapes and affected sutures in craniosynostosis. See text for details. (Reprinted with permission from Cohen MM Jr, MacLean<br />

RE: Anatomic, Genetic, Nosologic, Diagnostic, and Psychosocial Considerations. In: Cohen MM Jr, MacLean RE (eds), Craniosynostosis.<br />

Diagnosis, Evaluation, and Management, 2nd ed. New York, Oxford Univ Press, 2000; Ch 11, pp 119-143.)<br />

genesis and skeletal adjustments. The interlocking,<br />

peg-and-socket arrangement, allows osteogenesis to<br />

occur mainly at the bottom of the socket and point<br />

of the peg, for simultaneous jointing and growth of<br />

the bone against the suture. In contrast, the facial<br />

sutures, especially the zygomatic, maxillary, and<br />

palatine, are simply overlapping or sliding joints where<br />

the direction of bone growth tends to parallel the<br />

plane of the suture. This arrangement provides for<br />

adaptive adjustments to pressure in utero and early<br />

infancy.<br />

The intrinsic maxillary growth concept recognizes<br />

specific bone growth sites in the maxilla, with growth<br />

occurring mainly in a backward direction by osteogenesis<br />

on the retromaxillary surface. The nasal septum<br />

exerts forward pull on the maxilla at the insertion<br />

of the septopremaxillary ligament. 143,144 Mandibular<br />

growth is primarily through growth and elongation at<br />

the level of the ramus and subcondylar segments.<br />

Experimental studies indicate that condylar cartilage<br />

14<br />

growth is probably secondary to traction by the soft<br />

tissues within and attached to the mandible, such as<br />

the tongue and muscles of mastication. 55<br />

Pathogenesis of Craniosynostosis<br />

The pathogenesis of craniosynostosis is complex<br />

and probably multifactorial. Moss145 theorized that<br />

abnormal tensile forces are transmitted to the dura<br />

covering the brain from an anomalous cranial base<br />

through key ligamentous attachments, and this leads<br />

to craniosynostosis. This hypothesis fails to explain<br />

the coexistence of nonsyndromic craniosynostosis with<br />

a normal cranial base configuration.<br />

Cohen123,146 suggests craniosynostosis is either primary<br />

due to suture biology147 or secondary to<br />

another event such as in-utero compression of the<br />

cranium, 148,149 decompression of a hydrocephalus,<br />

150–152 inadequate intrinsic growth forces of the<br />

brain (microcephaly), hyperthyroidism, 153 ricketts,<br />

or shunted hydrocephalus.


Approximately 2% of cases of isolated craniosynostosis<br />

are inherited. 154 Cohen, Dauser, and Gorski 155<br />

documented three close relatives with delayed onset<br />

of exorbitism and midfacial retrusion thought to be<br />

consistent with a diagnosis of familial, nonsyndromic<br />

craniosynostosis. In contrast, a hereditary component<br />

has been identified in as many as 50% of<br />

syndromal craniosynostosis patients. 154<br />

The Role of the Dura in Craniosynostosis<br />

In cases of primary craniosynostosis the underlying<br />

dura mater acts locally to supply the overlying<br />

suture with osteogenic growth factors. Opperman in<br />

1993 showed the role of the dura in determining the<br />

fate of suture fusion, 156 and that these dural factors<br />

were soluble. 157 Hobar158,159 noted the importance<br />

of the dura in the regeneration of cranial bones in<br />

infants. Greenwald160,161 went further, finding that<br />

immature dura mater contained a subpopulation of<br />

osteoblast-like cells. Levine162 showed that the region<br />

of the dura was as important as the interaction<br />

between the suture and the dura. The overlying<br />

pericranium does not play a role in suture biology,<br />

according to Opperman. 163 Clearly the dura underlying<br />

the suture drives the timing of closure through<br />

osteoinductive growth factors<br />

Molecular Genetics in Craniosynostosis<br />

Elevated FGF-R2 was identified by Delezoide164 as the first real marker of prechondrogenic condensations.<br />

Mangasarian165 identified a tyrosine for cysteine<br />

substitution on the mutated FGF-R2 that creates<br />

an activated form, resulting in uncontrolled FGF<br />

signaling and premature suture closure.<br />

Most166 and Mehrara167 found that expression<br />

of basic fibroblast growth factor (bFGF) was<br />

increased in the dura beneath the suture prior to<br />

fusion and increased in the osteoblasts at the suture<br />

during fusion. Other growth factors have been<br />

found to be increased in prematurely fusing<br />

sutures: transforming growth factor B (TGF-<br />

B) 166,168–171 and bone morphogenic proteins<br />

(BMPs). 171 Research is now directed at inducing<br />

craniosynostosis in animal models by the application<br />

of exogenous FGFs to confirm their role in<br />

suture closure. 172 Genes whose mutations result<br />

in craniosynostosis syndromes are listed in Table<br />

4. 173,174<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

TABLE 4<br />

Genes Bearing Known Mutations<br />

for Craniosynostosis<br />

(Reprinted with permission from Cohen MM Jr: Discussion of<br />

“Differential expression of fibroblast growth factor receptors in<br />

human digital development suggests common pathogenesis in<br />

complex acrosyndactyly and craniosynostosis”, by Britto JA,<br />

Chan JCT, Evans RD, et al. Plast Reconstr Surg 107:1339, 2001.)<br />

In 1993 a mutation in the homeobox gene MSX2<br />

was identified in a single family with autosomal dominant<br />

craniosynostosis, now known as Boston-type<br />

craniosynostosis. 175 Recent evidence suggests that<br />

this mutation may exert its effect through BMP pathways<br />

176,177 and that MSX2 mutations may function<br />

together with FGF-R2. 178<br />

Crouzon syndrome has been extensively studied<br />

genetically and a mapped abnormality is noted on<br />

the long arm of chromosome 10. 179 Further analysis<br />

localized an FGF-R2 genetic mutation within this<br />

chromosome in subjects with Crouzon. 180<br />

In 1994 Muenke and colleagues 181 described a<br />

common mutation in the FGF-R1 gene as the cause<br />

of Pfeiffer’s syndrome. In 1995 Apert and Pfeiffer<br />

syndromes were also found to be related to FGF-R2<br />

mutations. 182–184 Mutations in the FGF-R2 gene now<br />

having been shown to be the cause of Jackson-Weiss<br />

syndrome, Crouzon syndrome, Pfeiffer syndrome, and<br />

Apert syndrome, these conditions should be seen as<br />

defined points along a spectrum of disease (Fig 12).<br />

FGF-R3 mutations are also implicated in isolated<br />

unicoronal craniosynostosis. 185 Current understanding<br />

allows a molecular diagnosis in all phenotypical<br />

cases of bicoronal synostosis. 186 Other genetic mutations<br />

have been associated with synostosis. The<br />

hedgehog family of homologs, including sonic hedgehog,<br />

play a role in vertebral embryogenesis. Recently<br />

15


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 12. Fibroblast growth factor<br />

mutations and their associated craniofacial<br />

anomalies. (Reprinted with<br />

permission from Cohen MM Jr: Discussion<br />

of “Differential expression of<br />

fibroblast growth factor receptors in<br />

human digital development suggests<br />

common pathogenesis in complex<br />

acrosyndactyly and craniosynostosis”,<br />

by Britto JA, Chan JCT, Evans<br />

RD, et al. Plast Reconstr Surg<br />

107:1339, 2001.)<br />

an association was made between craniofacial anomaly<br />

and sonic hedgehog target genes. 187–189 The TWIST<br />

gene locus regulates osteoblast differentiation190 and<br />

mutations are associated with Saethre-Chotzen syndrome,<br />

191–193 causing an up-regulation in FGF-R<br />

expression leading to premature oseoblast differentiation<br />

and cranial suture fusion. Rice194 proposes an<br />

integration between FGF and TWIST mutations in suture<br />

development and suggests that different mutations may<br />

work on the same pathway at different stages.<br />

Ting195,196 examined the molecular difference<br />

between fused and nonfused human coronal sutures<br />

and identified overexpresion of the Nell-1 gene, again<br />

related to premature osteoblast differentiation.<br />

It is not adherent tensile forces or an intrinsic property<br />

of the suture that determines suture biology, but<br />

rather a combination of dura-related genetic,<br />

molecular, and cellular factors that act individually<br />

or jointly in the development of craniosynostosis.<br />

Longaker197 provides a comprehensive review of current<br />

cranial suture research, concluding that gene<br />

therapy may offer targeted interventions that may<br />

alter synostosis onset and progression:<br />

Looking forward into the new millennium, one<br />

can imagine a time when major reconstructive<br />

surgery for craniosynostosis will no longer be<br />

necessary.<br />

Longaker (2001)<br />

16<br />

Indications for <strong>Surgery</strong><br />

Indications to proceed with surgical reconstruction<br />

of craniosynostosis include significant cranial and<br />

facial asymmetry, elevated intracranial pressure (ICP),<br />

and neuropsychologic disorders. In most cases of<br />

single suture craniosynostosis, “functional” indications<br />

(elevated ICP and treatable neuropsychologic disturbance)<br />

are not present, and the indication for surgery<br />

is the cranial/facial asymmetry, its degree and<br />

severity.<br />

The clinical symptoms of intracranial hypertension<br />

include headaches, irritability, and difficulty<br />

sleeping. Radiographic signs may include cortical<br />

thinning or a luckenschadel (beaten metal) appearance<br />

of the inner table of the skull. Unfortunately,<br />

clinical and radiographic signs are relatively late<br />

developments, and increased intracranial pressure<br />

may be present for some time before these signs<br />

arise. Because of this, most craniofacial surgeons<br />

advocate early treatment, especially of infants with<br />

multiple suture synostoses. 198<br />

Marchac and Renier136,199 found that intracranial<br />

pressure may be elevated in those with single suture<br />

synostosis, although it is more common with multiple<br />

suture involvement (13% and 42%, respectively). The<br />

mean ICP has been shown to decrease after cranial<br />

vault remodeling. Young children with craniosynos-


tosis usually have normal mental development, but<br />

the proportion of normal children decreases with<br />

age, particularly for children with multisuture synostosis<br />

displaying brachycephaly and oxycephaly. 200 On<br />

the basis of their studies, the authors recommend<br />

early surgery, as there is no reliable way to distinguish<br />

which infants will not have problems from craniosynostosis.<br />

201,202<br />

Kapp-Simon and colleagues, 203 on the other hand,<br />

longitudinally examined the mental development of<br />

infants before and after cranial release, and compared<br />

it with that of infants who were not surgically<br />

treated. The authors concluded that cranial release<br />

and reconstruction did not affect mental development<br />

either positively or negatively. Renier and<br />

Marchac, 204 in a commentary of the above paper,<br />

strongly dispute this finding and state that the number<br />

of patients in Kapp-Simon’s study was too small<br />

to warrant any conclusions.<br />

Subsequently Kapp-Simon 205 published her analysis<br />

of a series of 84 patients with single suture synostosis<br />

followed longitudinally for >1y and reported a mental<br />

retardation rate of 6.5%, which is 2–3X the normal.<br />

Almost half the children who were of school<br />

age displayed some type of learning disorder. More<br />

importantly, these results were independent of early<br />

surgical correction, discounting the hypothesis that<br />

early correction of craniosynostosis would improve<br />

mental function. Similarly, Virtanen 206 found that<br />

the neurocognitive performance of children with craniosynostosis<br />

did not reach that of matched normal<br />

controls, suggesting the impairment of brain function<br />

had already taken place in utero.<br />

Gault et al 207 attempted to correlate elevations of<br />

intracranial pressure with decreases in intracranial<br />

volume as measured by CT. They found no direct<br />

relationship between the two on measurement of<br />

104 children with craniosynostosis. 202 It appears that<br />

decreased intracranial volume alone is not adequate<br />

justification for surgery in cranisynostosis. 208<br />

Posnick and colleagues 209 demonstrated that true<br />

measurements of intracranial volume can be obtained<br />

indirectly using CT scans. Premature closure of either<br />

the sagittal or metopic suture does not result in<br />

diminished intracranial volume. This was confirmed<br />

in long-term follow-up by Polley, 210 who showed<br />

that craniofacial procedures can be relied upon to<br />

increase the intracranial volume. Polley also found<br />

that long-term normative intracranial volume was<br />

maintained postoperatively, and hypothesized that<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

although normal volumes are seen pre- and postoperatively<br />

in craniosynostosis, reconfiguration of the<br />

skull dimensions in the region of the synostosed suture<br />

may be beneficial.<br />

David et al 211 adds further support to this theory<br />

in a study of cerebral perfusion pre- and postoperatively,<br />

where they found cerebral perfusion defects<br />

preoperatively in the area of the fused suture that<br />

were corrected following surgery. In cases of complex<br />

craniosynostosis, abnormalities of venous drainage<br />

at the level of the skull base produce venous<br />

hypertension and subsequent raised ICP. 212 Most<br />

surgeons therefore operate on infants at age 6–9<br />

months, and even earlier in severe cases.<br />

Cohen 213 reviews the evidence and concludes that<br />

differences in methods of pressure measurement,<br />

patient selection, and the lack of normative data<br />

make interpretation of existing studies difficult. He<br />

suggests that a clinical awareness of the signs of raised<br />

ICP is essential, though in cases of moderate deformity<br />

and no signs of raised ICP, close follow-up is<br />

applicable.<br />

Neuropsychiatric disorders range from mild<br />

behavioral disturbances to overt mental retardation<br />

possibly secondary to cerebral compression. The<br />

abnormally shaped skull also imposes psychological<br />

considerations that can be severe and should not be<br />

underestimated. Barritt and associates 214 report that<br />

children with untreated scaphocephaly are teased<br />

and taunted at school for their head shape, which<br />

compounds their slow learning and poor motor skills.<br />

Arndt and others 215 and Pertschuk and Whitaker 216<br />

studied the psychosocial adjustments of children to<br />

the correction of a deformity by craniofacial surgery.<br />

The authors noted increased self-esteem and adaptive<br />

functioning along with a decrease in hyperactive<br />

behavior and inhibited attitude, peaking at 1 year<br />

postoperatively. Despite cosmetic improvements and<br />

lower anxiety levels, however, social interactions were<br />

not helped by the surgery. Likewise, Ousterhout<br />

and Vargervik 217 noted normalization of anthropometric<br />

points on CT scan of children who underwent<br />

Le Fort III and genioplasty because of craniosynostosis,<br />

but the degree of postoperative change did not<br />

equate with attractiveness.<br />

In another study, Barden and colleagues 218,219<br />

looked at changes in physical attractiveness as well as<br />

emotional and behavioral reactions of children before<br />

and after craniofacial surgery. Their findings suggest<br />

17


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

a positive effect of the surgery in terms of social<br />

interactions and the development of cognitive and<br />

emotional competence.<br />

ISOLATED CRANIOSYNOSTOSES<br />

Trigonocephaly<br />

Trigonocephaly accounts for 10–20% of all craniosynostosis<br />

cases, occurring in 1 in 2500–15000 newborns.<br />

220 Lajeunie221 found 237 cases of metopic<br />

synostosis for an incidence of 1 in 15000. The<br />

male:female ratio was 3.3:1 and 5.6% were familial.<br />

Infants with trigonocephaly have an easily visible<br />

and palpable midline frontal ridge that extends from<br />

the anterior fontanelle to the glabella. When viewed<br />

from above, the forehead resembles the keel of a<br />

boat. Sadove and coworkers222 note that depending<br />

on the timing and extent of premature suture closure,<br />

metopic synostosis can manifest as a spectrum<br />

of deformities ranging from an isolated midline forehead<br />

ridge to a keel-shaped frontal bony protruberance.<br />

The forehead deformity is accompanied<br />

by variable degrees of orbital hypotelorism, ethmoidal<br />

hypoplasia, and bitemporal narrowing. The<br />

intracranial volume of both frontal areas is often<br />

decreased, which results in compensatory overgrowth<br />

of both parietal areas. The frequency of elevated<br />

intracranial pressure is estimated to be approximately<br />

10%, 223 though it is likely that pressure is only an<br />

issue in the frontal lobes of those with severe deformity.<br />

Cognitive impairment is some series reaches<br />

33% 224 and correlates with severity of deformity, 225<br />

though it is likely to be an associated factor rather<br />

than a causative one.<br />

Posnick and associates226 evaluated the results of<br />

surgical intervention in metopic synostosis. They used<br />

CT scans to quantitatively record the deformities of<br />

uncorrected and corrected metopic synostosis,<br />

namely orbital hypotelorism, retruded lateral orbital<br />

rims, and narrow bitemporal width. Postoperative<br />

assessment confirmed that the anterior cranial vault<br />

and lateral orbital wall positions, which were initially<br />

dysmorphic, were corrected successfully and<br />

remained in good position despite subsequent calvarial<br />

growth (Fig 13). The orbital hypotelorism was<br />

improved but not totally corrected.<br />

Havlik et al227 examined 10 patients with severe<br />

trigonocephaly as defined by the central angle—the<br />

convergence of the two hemisupraorbital segments.<br />

18<br />

Fig 13. Left, a 10-month old boy with metopic synostosis and<br />

trigonocephaly. Right, after surgical correction with cranial vault<br />

and three-quarter orbital osteotomies. (Reprinted with permission<br />

from Posnick JC, Lin KY, Chen P, Armstrong D: Metopic<br />

synostosis: quantitative assessment of presenting deformity and<br />

surgical results based on CT scans. Plast Reconstr Surg 93:16,<br />

1994.)<br />

Preoperative angles were consistently in the vicinity<br />

of 110 °. In these cases they recommend expanding<br />

the supraorbital bar with interpositional bone grafts<br />

measuring 10–20mm, expanding the angle to<br />

approximately 145°, and thus widening the bitemporal<br />

narrowing (Fig 14). In milder cases they suggest<br />

that more conventional methods such as contour<br />

reduction or the floating forehead technique<br />

will be adequate.<br />

McCarthy and others 228 reviewed a 20-year experience<br />

with early surgery for isolated craniosynostosis.<br />

Of the 104 patients in their study, 29 had metopic<br />

suture synostosis. Orbital hypotelorism, initially<br />

apparent in 19, was significantly reduced postoperatively<br />

in 17 patients. In general, surgical correction<br />

of metopic synostosis was associated with the best<br />

aesthetic results of all the isolated craniosynostoses;<br />

only one patient required a second craniofacial procedure.<br />

McCarthy also followed 24 patients with<br />

metopic synostosis for whom surgical correction was<br />

not recommended. None of these patients demon-


Fig 14. Technique for correction of trigonocephaly showing two<br />

perspectives on the osteotomy sites (A & B, left) and after<br />

anterolateral transposition of the hemisupraorbital segment and<br />

bone grafting (A & B, right). (Reprinted with permission from<br />

Havlik RJ, Azurin DJ, Bartlett SP, Whitaker LA: Analysis and<br />

treatment of severe trigonocephaly. Plast Reconstr Surg 103:381,<br />

1999.)<br />

strated progression of the deformity, and 15 showed<br />

significant improvement in frontoorbital form. The<br />

authors believe that surgical correction is not justified<br />

in cases of metopic synostosis associated with only<br />

mild change in morphology and without evidence of<br />

functional disturbance. The risk of surgically induced<br />

complications, including failure of frontal sinus<br />

development, 229 outweighs the potential benefits of<br />

surgery.<br />

Scaphocephaly<br />

Sagittal synostosis, with its characteristic oblong<br />

calvarial shape (scaphocephaly), is the most common<br />

type of craniosynostosis. 230 The severity of the<br />

calvarial deformity varies from slight cranial elongation<br />

with sagittal ridging to extreme elongation with a<br />

large occipital shelf and pronounced frontal bossing.<br />

231 Persing, Jane, and Edgerton232 suggested that<br />

the deformity is not limited to the sagittal suture but<br />

also involves the base of the skull, which in part<br />

contributes to the degree of cranial dysmorphology.<br />

The optimal operative management of scaphocephaly<br />

is still debated. Multiple procedures are<br />

described in the literature. Strip craniectomy (Fig<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

15) as described by Ingraham 233 and its modifications<br />

234 are employed in cases of early diagnosis, but<br />

have produced unreliable results. 235–239 This may be<br />

due in part to recurrence of the synostosis 240 or<br />

deformation where rigid fixation is not employed.<br />

The “pi technique” as described by Jane 241 and its<br />

modifications 242 have been associated with elevation<br />

in intracranial pressure. 243 Techniques that employ<br />

wider stripping, such as total vertex craniectomy, 244<br />

produce a superior result 198 compared with strip<br />

craniectomy. 245 Kaiser 198 compared the postoperative<br />

results of midline craniectomy, bilateral<br />

parasagittal craniectomy, and total vertex craniectomy.<br />

Cases of vertex craniectomy of Epstein 244 all<br />

had normalization of the cephalic index, while the<br />

other two groups were successful only one-fourth to<br />

one-half of the time.<br />

The trend to more radical and extensive correction<br />

of the entire cranial vault 236,246–248 has led to the<br />

goal of actively correcting the cranial index and<br />

attaining an ideal cranial shape at the time of surgery.<br />

Postoperative passive correction is not expected.<br />

For severe cases of sagittal synostosis, Marchac and<br />

coworkers 249 perform frontocranial remodeling in one<br />

stage. The supraorbital bar is usually left in place<br />

and the upper forehead is reconstructed just above<br />

it. Posteriorly the occipital bone is moved forward<br />

and the vault is constructed of three or four bone<br />

segments cut transversely like parts of a barrel. These<br />

segments are rearranged to shorten the anteriorposterior<br />

distance, expand the transverse diameter,<br />

and lift up the retrocoronal depression.<br />

Older affected children who have not been operated<br />

on and children with failed previous attempts at<br />

scaphocephalic correction probably warrant a more<br />

aggressive approach consisting of total calvarial<br />

remodeling with anterior craniofacial reconstruction,<br />

temporoparietal widening, posterior remodeling, and<br />

anterior-posterior shortening if required. 250<br />

Plagiocephaly<br />

Deformational Plagiocephaly<br />

Mulliken251 and Huang and colleagues252 review<br />

the morphologic findings in posterior plagiocephaly<br />

and the differential diagnosis between positional and<br />

synostotic (Fig 16). In Huang’s prospective series of<br />

115 infants with posterior plagiocephaly, only one<br />

had lambdoid synostosis. 252<br />

19


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 15. Child with sagittal synostosis before (A-C) and after (D-F) surgical correction. (Reprinted with permission from Persing JA, Jane<br />

JA, Edgerton MT: Surgical Treatment of Craniosynostosis. In: Persing JA, Edgerton MT, Jane JA (eds), Scientific Foundations and<br />

Surgical Treatment of Craniosynostosis. Baltimore, Williams & Wilkins, 1989, p 191.)<br />

The importance of differentiating between<br />

deformational and synostotic plagiocephaly 251 is that<br />

conservative therapy will achieve results equivalent<br />

to those of surgery. 253,254 Deformational plagiocephaly<br />

is best treated with positioning, frequent head<br />

turning, and helmet therapy. 141,255 Vies, 142 reporting<br />

on a series of 105 patients treated with either head<br />

positioning or helmet therapy, noted faster and better<br />

results in the helmet group.<br />

Synostotic Plagiocephaly<br />

Posterior synostotic plagiocephaly is due to lambdoid<br />

synostosis and accounts for only 1% of occipital<br />

plagiocephaly cases. 256 In analyzing their series,<br />

Huang and colleagues139,252 found that in true lambdoid<br />

synostosis the contralateral posterior bossing<br />

occurred more laterally and superiorly in the parietal<br />

region; frontal bossing was not a striking feature, but<br />

20<br />

when it occurred it was contralateral rather than<br />

ipsilateral; and ipsilateral occipitomastoid bossing was<br />

consistently present in lambdoid synostosis, whereas<br />

it was conspicuously absent in deformational posterior<br />

plagiocephaly.<br />

The treatment implications are clear: Few patients<br />

with positional plagiocephaly actually require operative<br />

correction. Their skulls will remold satisfactorily<br />

with conservative measures (positioning of the child in<br />

the crib or helmet therapy). The surgical correction of<br />

posterior plagiocephaly caused by unilateral lambdoid<br />

synostosis is via posterior craniofacial reconstruction<br />

and remodeling, as recommended by Persing. 257<br />

Anterior synostostic plagiocephaly is due to unilateral<br />

coronal suture synostosis. Coronal synostosis<br />

may be either unilateral or bilateral. In a 21 year<br />

experience consisting of 116 patients with coronal<br />

synostosis, 47% were unicoronal, 9% were bicoronal<br />

without associated syndromes, 34% were bicoronal


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 16. Posterior plagiocephaly from positional molding (above) and unilambdoid synostosis (below). Arrows indicate vectors of<br />

compensatory growth. (Reprinted with permission from Huang MHS, Mouradian WE, Cohen SR, Gruss JS: The differential diagnosis of<br />

abnormal head shapes: separating craniosynostosis from positional deformities and normal variants. Cleft Palate Craniofac J 35:204, 1998.)<br />

associated with a syndrome, and the remaining 20%<br />

were associated with multiple suture synostosis. 258<br />

Unicoronal synostosis causes regional growth<br />

restriction and compensatory expansion of the neighboring<br />

tissues, producing overt frontoorbital<br />

dysmorphology. Characteristic deformities ipsilateral<br />

to the synostosis include flattening of the frontal bone<br />

and ipsilateral forehead, ipsilateral elevation-recession<br />

of the supraorbital rim, and narrowing and lateral<br />

deviation of the orbit, deviation of the nasal root<br />

towards the flattened side, and elevation of the ipsilateral<br />

ear. 259 On the contralateral side there is bulging<br />

of the frontal bone. 260,261 An AP radiograph usually<br />

demonstrates the characteristic harlequin eye<br />

deformity. Bruneteau and Mulliken 262 believe that<br />

physical examination focusing on the supraorbital<br />

rims, nasal root, ears, and malar eminences can easily<br />

distinguish between synostotic and deformational<br />

plagiocephaly. This distinction has obvious clinical<br />

implications, as synostotic plagiocephaly is the only<br />

group with strong surgical implications.<br />

Lo and colleagues 263 described the endocranial configuration<br />

in unilateral coronal synostosis as follows:<br />

• constriction of the ipsilateral anterior cranial fossa<br />

• deviation of the anterior fossa midline<br />

• elevation of the ipsilateral floor<br />

• straightening of the lesser sphenoid wing<br />

These features of the cranial base correlate with<br />

the orbital dysmorphology. The authors reviewed<br />

28 patients with unicoronal synostosis and noted more<br />

21


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

symmetrical bony orbits and orbital contents during<br />

the first year after cranioorbital surgery. They conclude<br />

that surgery for unicoronal synostosis (Fig 17)<br />

in infancy does not inhibit growth of orbital hard or<br />

soft tissues and seems to allow normalization of previously<br />

impaired growth. 265<br />

Fig 17. Anterior plagiocephaly and its surgical correction. A, site<br />

of anterior cranial vault and three-quarter orbital osteotomies. B,<br />

reshaping and fixation of bone segments after osteotomies.<br />

(Reprinted with permission from Posnick JC: Craniosynostosis:<br />

Surgical Management in Infancy. In: Bell WH (ed), Orthognathic<br />

and Reconstructive <strong>Surgery</strong>. Philadelphia, WB Saunders, 1992;<br />

vol 3, p 1837.)<br />

Bicoronal Synostosis<br />

Bicoronal synostosis produces an abnormal skull<br />

shape—brachycephaly. The calvarium is short<br />

anteroposteriorly, widened mediolaterally, and elongated<br />

vertically compared with normal subjects. The<br />

orbital rims are hypoplastic, the occipital region is flattened,<br />

the frontal and temporal bones are protruberant,<br />

and the anterior cranial base is foreshortened. 266<br />

Although the brachycephalic deformity is characteristic<br />

of bicoronal synostosis, morphologic variants have<br />

been recognized. 267<br />

22<br />

Marchac’s 268 series of 35 children included 20<br />

patients with ‘simple’ and ‘familial’ brachycephaly<br />

treated by his “floating forehead” technique. Only<br />

one patient (5%) required secondary frontal advancement.<br />

In his series of 22 consecutive patients with<br />

nonsyndromic bicoronal synostosis, Wagner 266 reports<br />

62% relapse in patients operated on at age 5 months<br />

or younger. These infants subsequently needed total<br />

reoperation. In contrast, only 11% of children operated<br />

on at age >6mo showed recurrence of the<br />

deformity. This observation is contrary to contemporary<br />

theory and the experimental and clinical<br />

impression of others, which indicate that normalization<br />

of calvarial shape is more likely after early surgery.<br />

269,270<br />

SYNDROMES WITH CRANIOSYNOSTOSIS<br />

Craniosynostosis is not a syndrome in itself but a<br />

sign of at least 150 different syndromes. 271 A<br />

multidisciplinary team involving craniofacial, ophthalmologic,<br />

and ear-nose-throat surgeons, geneticists,<br />

neuropsychologists, nutritionists, and social workers<br />

will allow total patient care. Priorities regarding<br />

intracranial pressure, airway obstruction, and globe<br />

exposure will dictate surgical priorities. Some of the<br />

more common syndromes with craniosynostosis as<br />

their predominant physical finding are described<br />

below (Table 5).<br />

Apert Syndrome<br />

Originally described by Wheaton in 1894, the<br />

condition is named after Apert, who in 1906<br />

described 4 cases. 272 The incidence of Apert syndrome<br />

is reported to be 1:100,000 to 1:160,000<br />

births. 273 Although transmission is autosomal dominant,<br />

occurrence is sporadic as new mutations with<br />

normal karyotype.<br />

The skull in Apert syndrome is brachycephalic. 274<br />

Facial features incude hypoplasia of the midface,<br />

hypertelorism, 275 strabismus, ocular muscle palsies, 276<br />

and slanting palpebral fissures. There is moderate to<br />

severe exorbitism, short zygomatic arches and, in the<br />

euryprosopic type of Apert, a prominent bregmatic<br />

bump. The fontanelles may be large and late in<br />

closing. The palate is narrow and either has a median<br />

groove or is cleft with a bifid uvula (Fig 18). The<br />

limbs show bony syndactyly (secondary to the FGF-R


mutations implicated in the craniosynostosis 173 ) with<br />

complete fusion of the fingers and a free thumb.<br />

The distal phalanx of the thumb is often broad.<br />

Cutaneous syndactyly of all toes may be either simple<br />

or complex. There is moderate to severe facial and<br />

upper extremity acne.<br />

Mental retardation is variously reported in association<br />

with Apert syndrome, but it is unclear how<br />

much of it is secondary to environmental influences.<br />

Certainly many patients with Apert syndrome attain<br />

a high level of mental function.<br />

TABLE 5<br />

Syndromic Craniosynostoses<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

(Reprinted with permission from Whitaker LA, Bartlett SP: <strong>Craniofacial</strong> <strong>Anomalies</strong>. In: Jurkiewicz MJ, Krizek TJ, Mathes SJ, Ariyan S (eds),<br />

<strong>Plastic</strong> <strong>Surgery</strong>. Principles and Practice. St Louis, CV Mosby, 1990. Vol 1, Ch 7.)<br />

Although head shape in Apert syndrome suggests<br />

bicoronal synostosis, a postmortem study of the craniofacial<br />

changes in the syndrome concluded that the<br />

pathology is not primarily due to craniosynostosis but<br />

rather stems from reduced growth potential of the<br />

cranial base, leading to premature fusion of the midline<br />

sutures from the occiput to the anterior nasal<br />

spine 273 and resulting in severe calvarial, maxillary,<br />

nasal, and mandibular abnormalities. Other autopsy<br />

reports 277 suggest that basisphenoid synchondrosis<br />

23


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 18. Child with Apert syndrome. A, B, clinical presentation. C, findings on 3-D reconstruction of CT scan. Note the synostotic coronal<br />

suture, the open fontanelle, and the midfacial hypoplasia. (Reprinted with permission from Buchman SR, Muraszko KM: Syndromic<br />

Craniosynostosis. In: Lin KY, Ogle RC, Jane JA (eds), <strong>Craniofacial</strong> <strong>Surgery</strong>. Science and Surgical Technique. Philadelphia, WB<br />

Saunders, 2002; Ch 18, pp 252-271.)<br />

may be the main factor in facial deformities through<br />

synostoses posteriorly and anteriorly to the vomer.<br />

For a comprehensive discussion of Apert syndrome,<br />

the reader is referred to the April 1991 issue of Clinics<br />

in <strong>Plastic</strong> <strong>Surgery</strong>, 278 which is devoted entirely to<br />

various aspects of the syndrome.<br />

Saethre-Chotzen Syndrome<br />

Saethre-Chotzen syndrome was first recognized<br />

as a discrete entity in 1931-32 by the physicians<br />

who gave their name to the syndrome. Saethre-<br />

Chotzen syndrome is characterized by a broad<br />

and variable pattern of craniofacial anomalies, frequently<br />

asymmetrical. Bicoronal synostosis, low<br />

hairline and upper eyelid ptosis characterize the<br />

syndrome. Intellect is normal. The midface is<br />

usually normal, which led Tessier273 to call it “upper<br />

Apert.” There is partial cutaneous or simple<br />

syndactyly, usually between the second and third<br />

fingers but sometimes involving the small finger<br />

too. Inheritance is autosomal dominant.<br />

Clauser279 reviewed the literature and found an<br />

incidence of 1 in 25000 to 1 in 50000. A mutation<br />

of the TWIST gene is identified in approximately<br />

68% of cases, and mutations of FGF-R2 and FGF-R3<br />

have been described.<br />

Pfeiffer Syndrome<br />

(Acrocephalosyndactyly Type V)<br />

Pfeiffer syndrome is characterized by broad thumbs<br />

and great toes. The severe midfacial hypoplasia<br />

24<br />

characteristic of Apert syndrome is present, but cranial<br />

vault malformations are not as extreme. Intellect<br />

is normal. Tessier 273 refers to Pfeiffer syndrome<br />

as a “low Apert.”<br />

A 1993 report 280 identified subgroups of Pfeiffer<br />

syndrome according to the apparently consistent patterns<br />

of phenotypic expression, including head shape.<br />

The classic syndrome (Cohen type I) consists of symmetrical<br />

bicoronal synostosis; all other sutures are<br />

normal. Cohen type II or cloverleaf skull is an<br />

expression of early, extensive multisuture synostosis.<br />

All affected subjects have multiple constricting rings<br />

of prematurely fused sutures, grossly foreshortened<br />

anterior and posterior cranial bases, and coincident<br />

hydrocephalus. Moore 281 states: “This early, extensive<br />

fusion of multiple suture rings in association with<br />

an intrinsic, or secondary, cranial base distortion promotes<br />

the development of hydrocephalus, raised<br />

intracranial pressure, calvarial vault thinning, and cranial<br />

lacunae. Despite early radical craniectomies<br />

and cranial vault reshaping in these patients, recurring<br />

fusion of the ‘neosutures’ is often rapid.”<br />

Cohen type III is intermediate in the pattern of<br />

extensive sutural involvement. In addition to the<br />

bicoronal synostosis, isolated fusion of other sutures<br />

is evident early. This becomes progressively more<br />

widespread with time. Cohen 280 describes patients<br />

with type III Pfeiffer syndrome as having severe ocular<br />

proptosis, an extremely short anterior cranial base,<br />

neurological compromise (hydrocephalus is common),<br />

and a poor prognosis with early death. All<br />

reported cases of type III Pfeiffer syndrome have<br />

been sporadic. 282


Pfeiffer syndrome is inherited as an autosomal<br />

dominant disorder. Muenke et al 283 and Kerr and<br />

colleagues 284 report an affected family in which some<br />

individuals have normal thumbs although several relatives<br />

do have hand anomalies (symphalangism). Not<br />

only is there phenotypic variability in Pfeiffer syndrome<br />

and other acrocephalic syndactylies, but recent<br />

molecular data indicate variable phenotypic expression<br />

for patients who have identical mutations. Point<br />

mutations in FGFR2 have been identified in Crouzon,<br />

Jackson-Weiss, and Apert syndromes in addition to<br />

sporadic cases of Pfeiffer syndrome. 285 In addition, a<br />

subset of Pfeiffer syndrome families have a common<br />

mutation in the FGFR1 gene.<br />

Carpenter Syndrome<br />

Carpenter syndrome was originally described in<br />

1901 by the man who lent it his name. Acrocephalopolysyndactyly<br />

or Carpenter syndrome consists<br />

of craniosynostosis, short fingers, soft tissue syndactyly,<br />

preaxial polydactyly, congenital heart disease,<br />

hypogenitalism, obesity, and umbilical hernia.<br />

As many as 75% of patients have some degree of<br />

intellectual impairment. 286<br />

The craniofacial anomalies are those of brachycephaly<br />

with variably severe synostosis of the coronal,<br />

sagittal, and lambdoid sutures, such that the<br />

calvarium is usually distorted and grossly asymmetrical,<br />

approaching the kleeblattschadel anomaly.<br />

Midfacial retrusion and dental malocclusion are less<br />

pronounced than in Apert, and syndactyly is only<br />

partial and usually simple. 273 This is one of the few<br />

craniofacial malformation syndromes that is inherited<br />

as an autosomal recessive.<br />

Poole287 cautions surgeons contemplating surgery<br />

on patients with Carpenter syndrome that they must<br />

beware of the venous and bony abnormalities attendant<br />

with this syndrome. The vascular anomalies<br />

may lead to excessive and rapid blood loss.<br />

Crouzon Syndrome<br />

The French neurosurgeon Crouzon described the<br />

disease that bears his name in 1912, at which time<br />

he listed four essential characteristics: exorbitism,<br />

retromaxillism, inframaxillism, and paradoxical<br />

retrogenia288 (Fig 19). The estimated incidence is 1<br />

in 25000 live births. 289 Inheritance is autosomal<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

dominant and occurrence is both sporadic and<br />

familial.<br />

Fig 19. Clinical presentation of a child with Crouzon syndrome.<br />

Note exorbitism and hypertelorism, retromaxillism and midfacial<br />

hypoplasia, pseudoprognathic mandible, and paradoxical retrogenia.<br />

(Reprinted with permission from Buchman SR, Muraszko KM:<br />

Syndromic Craniosynostosis. In: Lin KY, Ogle RC, Jane JA (eds),<br />

<strong>Craniofacial</strong> <strong>Surgery</strong>. Science and Surgical Technique. Philadelphia,<br />

WB Saunders, 2002; Ch 18, pp 252-271.)<br />

There are many clinical variations of Crouzon disease,<br />

often with one or more of the “typical” signs<br />

missing. 290 The shape of the skull alone does not<br />

differentiate Apert syndrome from Crouzon disease,<br />

although Crouzon shows less severe deformity.<br />

Crouzon is characterized by recession of the frontal<br />

bone and supraorbital rim, retrusion of the facial<br />

mass, exorbitism with proptosis, and hypoplasia of<br />

the infraorbital rim. Unlike Apert syndrome,<br />

hypertelorism and a bregmatic bump may not be<br />

evident, the nose does not always deviate, and the<br />

orbital deformities tend to be symmetrical. The orbits<br />

are quite shallow, with hypoplastic and flat infraorbital<br />

rims, distention of the eyelids, palpebral fissures<br />

slanted downward, exophoria, and divergent strabismus;<br />

50% of patients with Crouzon disease have<br />

hyperactivity of the inferior oblique muscles. Crouzon<br />

patients usually develop normal intelligence and limb<br />

anomalies are absent. The mandible in Crouzon<br />

disease, as in Apert, is considerably shorter than normal,<br />

producing a distinctive ramus/body length ratio,<br />

particularly in older patients. 291<br />

25


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Cephalometric analysis of patients with Crouzon<br />

disease shows a direct correlation between the altered<br />

facial morphology and abnormalities of the cranial<br />

base. The exorbitism is a reflection of vertical sloping<br />

of the anterior cranial fossa, while the midfacial retrusion<br />

can be traced to angulation of the cranial base<br />

flexure. 292<br />

Proudman and colleagues 293 reviewed 59 patients<br />

with Crouzon disease to determine the noncraniofacial<br />

manifestations. They conclude that<br />

Crouzon syndrome is not just a craniofacial deformity<br />

but “a dysplastic condition that can affect other<br />

areas of the bone and chondral growth, such as the<br />

spine and elbows. Ligaments and soft tissues may<br />

also be involved.” Cervical spine anomalies (40%),<br />

stylohyoid calcification (50%), elbow anomalies (18%),<br />

minor hand deformities (10%), other musculoskeletal<br />

anomalies (7%), and visceral anomalies (7%) were<br />

all present.<br />

Kreiborg 294 traces the craniofacial growth, clinical<br />

findings, craniofacial morphology, occlusion, and<br />

other aspects of dysmorphology in 61 patients with<br />

Crouzon syndrome before treatment. Extensive statistical<br />

data on adult skeletal dimensions are given, as<br />

well as a comprehensive bibliography. Later, Kreiborg<br />

and colleagues 295 used 3D CT scans to compare the<br />

anatomy of the calvarial cranial base in Apert and<br />

Crouzon syndromes. Their findings suggest that the<br />

conditions are very different in cranial development.<br />

Cartilage abnormalities, particularly those of the<br />

anterior cranial base, play a major role in cranial<br />

development in Apert syndrome, whereas the primary<br />

abnormality in Crouzon appears to be premature<br />

fusion of sutures and synchondrosis. The authors<br />

advocate early surgery in both groups of patients,<br />

but for different reasons: In Apert syndrome, early<br />

surgery is indicated to reduce further dysmorphic<br />

growth changes in the calvarial cranial base; in<br />

Crouzon syndrome, early surgery can prevent or<br />

relieve elevated intracranial pressure.<br />

Posnick and colleagues 296 reviewed their experience<br />

in 14 children with Crouzon syndrome who<br />

presented with bicoronal synostosis. Measurements<br />

of the cranioorbitozygomatic region taken from CT<br />

scans done pre- and postoperatively served to document<br />

the results of surgical correction. Early surgical<br />

attempts to decompress and reshape the cranioorbital<br />

regions may limit the effects of increased intracranial<br />

pressure but do not correct the deformity, as judged<br />

by CT scans. Although the Crouzon deformity did<br />

26<br />

not worsen after surgery, the measurements remained<br />

far from normal. Perhaps more importantly, the<br />

maxillary and mandibular deformities were not<br />

addressed and the vertical dimension of the face was<br />

unchanged.<br />

In their review of syndromic craniosynostoses<br />

treated at NYU, McCarthy and associates 269 note<br />

disappointing aesthetic and functional results of surgery<br />

compared with cases of isolated craniosynostosis.<br />

The incidence of major secondary procedures,<br />

peri- and postoperative complications, hydrocephalus,<br />

shunts, and seizures was significantly increased<br />

in syndromic patients. Frequent problems included<br />

increased intracranial pressure, airway obstruction,<br />

and recurrent turricephaly or cranial vault maldevelopment.<br />

Moreover, early frontoorbital advancementremodeling<br />

“failed to promote midface development.”<br />

Craniofrontonasal Dysplasia<br />

Craniofrontonasal dysplasia (CFND) is a rare<br />

familial craniofacial disorder first described by<br />

Cohen. 297 The syndrome combines coronal craniosynostosis<br />

and frontonasal dysplasia with a variety of<br />

extracranial abnormalities. The coronal synostosis<br />

results in brachycephaly (usually asymmetrical) and<br />

frontal bossing, while the frontonasal “dysplasia”<br />

manifests as hypertelorism and a broad nose, frequently<br />

with a bifid nasal tip. Other features are<br />

curly hair, grooved nails, cleft lip and palate, high<br />

arched palate, strabismus, shoulder and hip girdle<br />

anomalies, soft-tissue syndactyly of fingers and toes,<br />

and a broad great toe. 298<br />

Orr and colleagues299 reviewed their experience<br />

with craniofrontonasal dysplasia in 10 female patients.<br />

Male patients seem to have a milder phenotype.<br />

The precise mode of genetic transmission is unclear,<br />

but maternal transmission to daughters and sons has<br />

been reported. Affected males have passed the condition<br />

to 100% of daughters in some published pedigrees,<br />

but male to male transmission is not known to<br />

occur. Treatment is in two stages, one for correction<br />

of the craniosynostosis and the other to deal with the<br />

hypertelorism. Six patients had facial bipartition and<br />

3 patients had combined intra- and extracranial<br />

periorbital osteotomies, resection of excess midline<br />

or paramedian bony tissue and ethmoid sinuses, and<br />

medial translocation of the orbits.


Associated <strong>Anomalies</strong><br />

Although cervical spine anomalies are not usually<br />

mentioned in association with craniosynostosis,<br />

intervertebral fusion has been documented in 71%<br />

of children with Apert syndrome, in 38% of those<br />

with Crouzon disease, and in 30% of those with<br />

Pfeiffer syndrome. 300 Fusions are most often isolated<br />

and involve complex C5-6 lesions in Apert syndrome<br />

and the upper-level cervical vertebrae in Pfeiffer and<br />

Crouzon patients. Affected children may have limited<br />

range of cervical motion, which has implications<br />

for airway management during surgery. 300<br />

III — ATROPHY/HYPOPLASIA<br />

Romberg Disease<br />

(Progressive Hemifacial Atrophy)<br />

Romberg disease was first described by Parry301 in<br />

1825 and later by Romberg302 in 1946. Eulenberg303 coined the term “progressive facial hemiatrophy” in<br />

1871. The disease commences usually in the first or<br />

second decade of life and is more common in girls<br />

than boys by a 1.5:1 ratio. 304 The atrophy is unilateral<br />

in 95% of cases and affects either side of the<br />

face with equal frequency.<br />

The etiology of the disorder is unknown, although<br />

many theories for its pathogenesis have been proposed.<br />

Foremost among these are infection, 305<br />

trigeminal peripheral neuritis, 306 scleroderma, 307 and<br />

cervical sympathetic loss. 308<br />

The condition manifests as progressive hemifacial<br />

atrophy of skin, soft tissue, and bone. Pensler and<br />

colleagues308 evaluated 41 patients and noted that all<br />

atrophic changes began in a localized area and progressed<br />

at a variable rate within the dermatome of<br />

one or more branches of the ipsilateral fifth cranial<br />

nerve. The average age at inception of the disease<br />

was 8.8 years. The main period of progression was<br />

8.9 ± 6 years. In 26 patients with skeletal involvement,<br />

the mean age of onset was 5.4 years vs 15.4<br />

years for 15 patients without skeletal involvement.<br />

No correlation could be established between the<br />

severity of soft-tissue deformity and the age of onset.<br />

Tissue from 6 patients who had ultrastructural<br />

analysis revealed a lymphocytic neurovasculitis with<br />

striking abnormalities of the vascular endothelium<br />

and basement membrane. The alterations of the<br />

vascular basal lamina in lymphocytic neurovasculitis<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

appears to reflect chronic vascular damage with repeat<br />

attempts at endothelial cell regeneration.<br />

Moore and colleagues 309 noted 50% of patients<br />

with Romberg disease had the classic early sign of<br />

coup de sabre, reflecting soft-tissue involvement in<br />

the upper face (frontal and maxillary dermatomes).<br />

In the presence of prolonged active disease, the softtissue<br />

atrophy extended to involve the whole hemiface.<br />

Late-onset disease appears to be characterized by<br />

soft-tissue atrophy in the lower face. Bony hypoplasia<br />

in the mid and lower face was most common.<br />

Involvement of the frontal region was relatively infrequent.<br />

The derangement of the craniofacial skeleton is<br />

unlikely to be solely due to an isolated intrinsic process.<br />

Moore et al 309 surmise that restriction of the<br />

abnormal soft-tissue envelope undoubtedly compounds<br />

any primary skeletal growth disturbance. If<br />

the disease involves bone, it likely exerts its effect on<br />

the craniofacial skeleton only during periods of facial<br />

growth acceleration.<br />

Treatment involves 3D reconstruction of all softtissue<br />

and skeletal disturbances. <strong>Surgery</strong> is usually<br />

undertaken at least 1 year after photographic records<br />

show no further loss of volume. Greater omentum<br />

free flaps have been described by Jurkiewicz and<br />

Nahai, 310 who note problems with lack of structural<br />

strength and gravitational descent.<br />

Inigo and colleagues 311 review their experience<br />

with dermis-fat free flaps in 35 patients with Romberg<br />

disease. They used the groin free flap in 33<br />

patients and a scapular flap in 3 patients in a twostage<br />

procedure consisting of transfer of the free<br />

flap and defatting and repositioning 6 months later.<br />

Adjuvant procedures included temporal fascial flaps<br />

for the frontal regions and cartilage grafts in the<br />

piriform fossa. The chin was corrected by either<br />

sliding osteotomies for projections of >1cm and<br />

by alloplastic implants for projections of


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

placing the dermis outward. Harashina and Fujino314 argue that placing the dermis side down should reduce<br />

gravitational sagging, one of the major problems with<br />

the reconstruction in this situation. The abdominal<br />

flap, a variation of the groin flap based on the inferior<br />

epigastric vessels, has also been reported. 310,315<br />

Koshima et al316 describe their experience with the<br />

deep inferior epigastric perforator flap (DIEP).<br />

Mordick and colleagues317 compared the outcome<br />

of reconstruction with dermal fat grafts (8) vs vascularized<br />

tissue transfers (8) and and note that vascularized<br />

transfers provide better augmentation,<br />

although dermal fat grafts gave a satisfactory result in<br />

mild to moderate defects. Dermal fat grafting is a<br />

shorter procedure requiring less anesthetic time, less<br />

technical expertise and support, and shorter hospital<br />

stays than microsurgical transfers. Free flaps containing<br />

adipose tissue appear to increase in volume during<br />

growth and may also increase with weight gain.<br />

As their series progressed, the authors opted increasingly<br />

for augmentation with the scapular flap because<br />

of its large-caliber vessels and long pedicle. Their<br />

patients averaged 3.3 procedures for final correction.<br />

Upton and colleagues318 report their experience<br />

with scapular flaps for cheek reconstruction in 28<br />

patients, 5 of whom had Romberg disease. The<br />

major advantages of the scapular flap were a constant<br />

proximal vascular anatomy, long vascular pedicle<br />

with large-caliber vessels, large amount of available<br />

tissue (including bone), relatively hairless skin in most<br />

people, and minimal or no functional problems at<br />

the donor site. Disadvantages included the lack of<br />

sensation, poor external cheek skin color match, and<br />

a predictably widened scar at the donor site. The<br />

study produced the following observations:<br />

• deficiency of the malar prominence cannot always<br />

be corrected with soft tissue alone and often needs<br />

bone grafting or alloplastic implant;<br />

• when long fascial extensions are used for correction,<br />

they must be transferred across the midline<br />

of the upper and lower lips;<br />

• the area most consistently undercorrected is the<br />

medial portion of the deficient upper and lower lip;<br />

• the area most consistently overcorrected overlies<br />

the mandibular body and ramus.<br />

Longaker and Siebert 319 reported their experience<br />

with 15 cases of Romberg disease representing 16<br />

28<br />

free tissue transfers. Deepithelialized, extended<br />

parascapular flaps with large fascial extensions of the<br />

dorsal thoracic fascia were used for reconstruction.<br />

The fascia can be folded into variable thicknesses to<br />

correct subtle contour defects of the upper lip, medial<br />

canthus, eyelids, and other facial features traditionally<br />

difficult to reconstruct. These extensions can be placed<br />

easily across the midline to interdigitate with normal<br />

tissues at the boundary of the facial deformity.<br />

Past treatment methods have included silicone fluid<br />

injections, 320 which are contraindicated because of<br />

long term complications, and injections of lipoaspirated<br />

fat, which have met with mixed results. 321<br />

Autologous fat injections certainly have a place in<br />

the correction of mild contour irregularities. Muscular<br />

atrophy of pedicled or free muscle flaps present a<br />

problem in calculating the final volume needed for<br />

the repair. 322<br />

IV — NEOPLASIA/HYPERPLASIA<br />

<strong>Craniofacial</strong> Tumors<br />

Intracranial neoplasms may be of many different<br />

types, such as fibrous dysplasia, simple osteoma,<br />

benign angiofibroma, neural tumor—specifically<br />

meningioma with extracranial erosion, encapsulated<br />

neurofibroma, schwannoma, or neurilemmoma323–325<br />

—cutaneous carcinoma, osteogenic<br />

sarcoma, and rhabdomyosarcoma.<br />

The first major offshoot from craniofacial surgery<br />

was the application of craniofacial techniques to the<br />

ablation of intracranial tumors. As clinical experience<br />

mounted, the following principles for the surgical<br />

management of craniofacial neoplasms evolved:<br />

• It is now possible to resect lesions that were previously<br />

considered unresectable. 326<br />

• Tumor position should be related to the base of<br />

the skull. 327<br />

• Tumor resection demands strict adherence to<br />

guidelines designed for tumor location, cell type,<br />

and stage, as well as the concept of complete “en<br />

bloc” excision. This latter is possible through<br />

regional orbitotomies for confined orbital tumors,<br />

bifrontal craniotomy, and facial incisions (eg,<br />

Weber-Ferguson) where necessary.<br />

• There is significant risk of losing the frontal bone<br />

flap to infection. 328,329


• The transfacial approach330 may reduce the frequency<br />

of infectious complications.<br />

• It is essential to prevent communication between<br />

the sinuses or nasal cavity and the meninges or<br />

intracranial dead space. The best way to achieve<br />

this is by interposing thin, vascularized tissue, such<br />

as a pedicled paracranial flap for very small defects,<br />

a galea frontalis flap for anterior defects, 331 and a<br />

temporalis muscle332 or temporoparietalis fascial<br />

flap for lateral and central defects.<br />

• Free flaps may be used not only to isolate and<br />

protect vital structures, but also to bring large<br />

amounts of soft-tissue bulk for contouring. Free<br />

flaps may be transferred secondarily to deal with<br />

complications, but probably should be raised prophylactically<br />

at the time of the ablative procedure.<br />

FIBROUS DYSPLASIA<br />

The most common osseous craniofacial tumor<br />

encountered by plastic surgeons is fibrous dysplasia.<br />

335 Fibrous dysplasia is an uncommon, nonneoplastic,<br />

benign disease of bone that was was first<br />

described by von Recklinghausen in 1891. 336 The<br />

pathogenesis of fibrous dysplasia involves abnormal<br />

activity of the bone-forming mesenchyme with an<br />

arrest of bone maturation in the woven bone stage,<br />

forming irregularly shaped trabecula. Mutations of<br />

signaling protein and increased IL-6 levels have been<br />

implicated in the process. 337 The condition is usually<br />

progressive until age 30, and reports of progression<br />

well into adulthood are not uncommon. 338<br />

Fibrous dysplasia in the cranioorbital area tends to<br />

be more osseous than fibrous dysplasia of other sites.<br />

Two patterns of presentation predominate: the<br />

monostotic form, with single-bone involvement, and<br />

the polyostotic variety, which may be associated with<br />

abnormal skin pigmentation, premature sexual<br />

development, and hyperthyroidism (Albright syndrome).<br />

The monostotic form is approximately 4X<br />

more common than the polyostotic form and<br />

approximately 30X more frequent than the complete<br />

Albright syndrome. The monostotic form commonly<br />

involves the ribs, femur, tibia, cranium, maxilla,<br />

and mandible. The most commonly affected<br />

bones in the cranium are the frontal and sphenoid<br />

bone; in the face, the maxilla338,339 (Fig 20).<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Posnick339 lists the keys to management of fibrous<br />

dysplasia, namely<br />

• accurate diagnosis<br />

• radiographic assessment<br />

• clinical evaluation<br />

• planning of interventions<br />

• long term follow-up<br />

Deformation caused by fibrous dysplasia of the<br />

anterior skull base can affect the architecture of<br />

the orbits and paranasal sinus, causing orbital displacement<br />

and exophthalmos. 340,341 In craniofacial<br />

fibrous dysplasia, the symptoms are varied and<br />

related to tumor mass, and may include facial pain<br />

and swelling, headaches, anosmia, deafness, blindness,<br />

malocclusion with displacement of teeth,<br />

diplopia, proptosis, and orbital dystopia. Cranial<br />

nerve palsies including optic nerve compression<br />

are not uncommon. 342 Eye symptoms may include<br />

extraocular muscle palsy and trigeminal neuralgia<br />

secondary to compression of the third and fifth<br />

cranial nerves. Orbital apex compression can initiate<br />

the cascade of ophthalmic venous engorgement,<br />

optic nerve atrophy, and loss of vision. Sphenoid<br />

body involvement can cause blindness as a<br />

result of compression of the optic nerve between<br />

the chiasm and optic foramen. Other minor ophthalmic<br />

symptoms include epiphora produced by<br />

occlusion of the lacrimal duct and visible external<br />

lid deformities. 343<br />

Malignant degeneration occurs in approximately<br />

0.5% of cases. 344 Clinical signs of malignancy consist<br />

of a rapid increase in size, pain, intralesional necrosis,<br />

bleeding, and elevation of serum alkaline phosphatase<br />

levels. The most common transformation is<br />

to osteogenic sarcoma, but fibrosarcoma and chondrosarcoma<br />

are also seen. The mean interval from<br />

diagnosis of fibrous dysplasia to evidence of malignancy<br />

is 13.5 years. 343 The polyostotic form of the<br />

disease has a higher incidence of malignant degeneration,<br />

although in the craniofacial region the<br />

monostotic form is more common. Malignant<br />

degeneration can occur spontaneously or following<br />

radiotherapy.<br />

Cherubism is a genetic disorder of the mandible<br />

and maxilla of the giant-cell type. It is strictly a selflimiting<br />

disease of children that regresses without surgery<br />

and leaves no deformity. 345<br />

29


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 24. A 5-year-old girl with polyostotic fibrous dysplasia and massive involvement of the maxilla and mandible bilaterally. A, C, D, before<br />

operation. B, 10 days after radical maxillary and mandibular debulking. (Reprinted with permission from Posnick JC, Hughes CA, Milmoe<br />

G, et al: Polyostotic fibrous dysplasia: an unusual presentation in childhood. J Oral Maxillofac Surg 54:1458,1996.)<br />

In the past, conservative treatment of fibrous dysplasia<br />

was preferred, occasionally with bonecontouring<br />

procedures. Spontaneous involution will<br />

not occur, however, and early surgical intervention<br />

when symptoms are just beginning may avoid extensive<br />

resection later. 346 When clinically feasible, the<br />

mainstay of therapy is complete or near-complete<br />

resection and reconstruction with normal autogenous<br />

bone.<br />

In 1972 Derome 347 pioneered the concept of total<br />

excision and immediate reconstruction of bone<br />

tumors, including 4 cases of fibrous dysplasia. Chen<br />

and colleagues 348 discussed the benefits and risks of<br />

prophylactic optic nerve decompression before symptoms<br />

develop. The decision for surgery in these<br />

cases is made solely on the basis of CT findings of<br />

encroachment of the optic canal by fibrous dyspla-<br />

30<br />

sia. The primary justification for prophylactic<br />

decompression is the speed with which visual deterioration<br />

can occur and the brief interval before it<br />

becomes permanent. Among reports of sudden loss<br />

of vision there are several that attributed the cause of<br />

blindness to hemorrhage or mucocele secondary to<br />

fibrous dysplasia. 349 In their own study of 18 patients<br />

with clinical or radiological evidence of optic canal<br />

involvement, Chen and associates 348 report 6 patients<br />

(33%) had loss of effective vision in the involved eye.<br />

From this experience and a review of the literature,<br />

the authors developed an algorithm for decompression<br />

of the optic nerve in fibrous dysplasia. Absolute<br />

indications for decompression are<br />

• progressive gradual visual loss<br />

• within 1 week of sudden visual loss


Relative indications for decompression are<br />

• patients presenting within 2–3 weeks of rapid visual<br />

loss<br />

• children or adolescents with no visual loss but<br />

radiographic evidence of optic canal reduction,<br />

since they are likely to have progressive growth of<br />

fibrous dysplasia<br />

• patients with no visual loss who have radiographic<br />

evidence of optic canal reduction and continuing,<br />

active fibrous dysplasia.<br />

Reconstruction after resection is typically immediate.<br />

Edgerton and colleagues336 described the use of<br />

the dysplastic bone as grafts, which following resection<br />

were contoured, thinned, and replaced. These<br />

grafts of dysplastic bone seem to function similarly to<br />

normal autogenous grafts and lack the potential for<br />

gradual recurrence of bone thickening frequently seen<br />

with in situ bone contouring. Autoclaving350 and cryotherapy351,352<br />

have been proposed to destroy the cellular<br />

elements and yet preserve the mineral matrix<br />

prior to reinsertion.<br />

Chen and Noordhoff353 analyzed their experience<br />

with the treatment of 28 patients with fibrous dysplasia.<br />

The authors outline a protocol for surgery (Table<br />

6) that includes<br />

1. total excision of dysplastic bone of the frontoorbital,<br />

zygomatic, and upper maxillary region<br />

and primary reconstruction with bone grafts;<br />

2. conservative excision of bone from under the hairbearing<br />

skull, central cranial base, and toothbearing<br />

regions; and<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

3. optic canal decompression in patients with orbital<br />

involvement and decreasing visual acuity.<br />

In a follow-up averaging 5.3 years, they find no<br />

recurrence or invasion of fibrous dysplasia into the<br />

grafted bone, but 5 of 19 patients treated with<br />

reduction of alveolar dysplasia had recurrence of the<br />

deformity that necessitated reshaping. One patient<br />

with recurrent mandibular fibrous dysplasia was successfully<br />

treated with hemimandibulectomy and mandibular<br />

reconstruction with vascularized bone graft.<br />

Hansen-Knarhoy and Poole 354 remark on the difficulty<br />

differentiating fibrous dysplasia from<br />

intraosseous meningioma around the orbital apex.<br />

From a review of their files, they were able to summarize<br />

the differences as follows:<br />

• Fibrous dysplasia commences in childhood and<br />

early adolescence while meningiomas are diseases<br />

of middleage.<br />

• Visual symptoms are prominent in meningioma<br />

cases and uncommon in fibrous dysplasia.<br />

• Proptosis is much more marked than orbital dystopia<br />

in the meningioma group while the reverse<br />

is true in fibrous dysplasia.<br />

• Fibrous dysplasia patients have extensive frontal<br />

bone involvement that is clinically obvious. There<br />

is no evidence of this in meningioma patients.<br />

• Pain is present in both groups and is not a useful<br />

discriminating feature.<br />

Most fibrous dysplasia lesions can be only partly<br />

excised and some are unresectable, arguing for treat-<br />

TABLE 6<br />

Treatment Protocol Based on Location, as Proposed by Chen and Noordhoff<br />

(Reprinted with permission from Chen Y-R, Noordhoff MS: Treatment of craniomaxillofacial fibrous dysplasia: How early and how<br />

extensive? Plast Reconstr Surg 86:835, 1990.)<br />

31


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

ment modalities other than surgery. No medical<br />

treatment is available to cure or definitively halt the<br />

progression of fibrous dysplasia. Reports of promising<br />

responses to systemic treatments with biphosphonate<br />

pamidronate await further studies before conclusions<br />

can be drawn. 355 Clark and Hobar 356<br />

implanted fibrous dysplasia cells in a nude mouse<br />

model and successfully decreased the size of the<br />

tumor with tamoxifen administration.<br />

NEUROFIBROMATOSIS<br />

Neurofibromatosis is a hereditary condition<br />

occurring in 1 in 3000 live births. Approximately<br />

50% of patients have a positive family history of neurofibromatosis.<br />

The term neurofibromatosis is applied<br />

to two clinical genetic disorders. The first, neurofibromatosis<br />

type 1 (NF1), also known as von<br />

Recklinghausen disease, is more common. The second,<br />

neurofibromatosis type 2 (NF2), is a rarer condition<br />

and is known as bilateral acoustic neurofibromatosis.<br />

The two disorders have very distinctive clinical<br />

manifestations and some overlapping features.<br />

Mutation of genes on two different chromosomes<br />

are at the origin of the two disorders. The gene for<br />

NF1 is located on the long arm of chromosome 17,<br />

while the gene for NF2 is located on the long arm of<br />

chromosome 22. 357 Transmission is autosomal dominant,<br />

with variable penetrance.<br />

Neurofibromatosis is a benign tumor of neuroectodermal<br />

origin with diffusion to skin, subcutaneous<br />

tissue, and bone (Fig 21). Tumor growth is slow and<br />

irregular and not accelerated by surgical intervention.<br />

Sarcomatous degeneration is rare.<br />

Surgical care often produces less than complete<br />

correction, with some further progression over<br />

time. 358,359 Poole, 358 in a series of 11 patients, stresses<br />

the high complication rate and modest improvement<br />

in appearance that should be expected. The best<br />

cosmetic results are obtained in patients whose eye<br />

can be removed and a satisfactory bed created for<br />

an orbital prosthesis. Patients who have a seeing eye<br />

and wish to retain it should undergo a two-stage<br />

procedure with conservative debulking of intraorbital<br />

soft-tissue.<br />

Krastanova-Lolov and Hamza357 reviewed their<br />

experience with 14 cases of cranioorbital neurofibromatosis.<br />

They note the partial or complete<br />

absence of the greater wing of the sphenoid is<br />

responsible for enlargement of the sphenoidal fis-<br />

32<br />

Fig 21. Patient showing clinical evidence of advanced neurofibroma.<br />

Photo courtesy of PC Hobar MD.<br />

sure, with a consequent defect in the posterior wall<br />

of the orbit. Brain tissue, generally the temporal<br />

lobe, may herniate into the orbit, further increasing<br />

exorbitism and causing pulsation of the eye. The<br />

orbit is enlarged, with hypoplasia of the supraorbital<br />

and infraorbital rims and the zygomatic arch. When<br />

the globe is involved with the neurofibroma there<br />

may be buphthalmos, severely diminished visual acuity,<br />

and even blindness. Associated symptoms are<br />

irritation, pain in the eye, and moderate to severe<br />

epiphora. Enophthalmos may occur from enlargement<br />

of the inferior orbital fissure, which allows the<br />

orbital contents to prolapse into the infratemporal<br />

fossa.<br />

Snyder 360 describes the experience of the Australian<br />

<strong>Craniofacial</strong> Unit in correcting 14 cases of orbital<br />

neurofibromatosis. Of note is the finding of resorption<br />

of the bone graft used to reconstruct the greater<br />

wing of the sphenoid in four cases, which led to recurrence<br />

of globe pulsation. Subsequently the author<br />

began using titanium mesh in the reconstruction.<br />

Jackson 361 presents his experience with orbitotemporal<br />

neurofibromatosis in 24 patients and provides<br />

a classification and treatment scheme. He<br />

groups the condition into three categories, each of<br />

which requires a different treatment: (1) orbital softtissue<br />

involvement with a seeing eye; (2) orbital softtissue<br />

and significant bone involvement with a seeing<br />

eye; and (3) orbital soft-tissue and significant bone<br />

involvement with a blind or absent eye. The author


traces the patients’ course over a maximum of 12<br />

years of recurrence-free follow-up.<br />

V — UNCLASSIFIED<br />

A few rare anomalies do not fit into the first four<br />

categories and are best placed in this last category.<br />

They should be described based an the organ or<br />

organs involved. Examples include aglossia or macroglossia,<br />

anotia,and ocular anomalies such as<br />

epicanthal folds. 10<br />

CRANIOMAXILLOFACIAL SURGERY<br />

HISTORY<br />

The origin of craniofacial surgery can be traced as<br />

far back as 1890, when Lannelongue and Lane performed<br />

the first craniotomies, 362,363 but it was not<br />

until the First and Second World Wars that numerous<br />

battle casualties stimulated the development of<br />

techniques for the replacement of missing bone and<br />

soft-tissue of the face, and set the stage for attempts<br />

in the 1940s and ’50s to correct congenital facial<br />

deformities.<br />

Waterhouse 364 reviews the history of craniofacial<br />

surgery with particular emphasis on the contributions<br />

by Le Fort, Virchow, Gillies, and Tessier. In<br />

1951 Gillies and Harrison 365 reported the first successful<br />

Le Fort III advancement osteotomy for correction<br />

of midfacial retrusion in a patient with<br />

Crouzon syndrome. The operation was technically<br />

very complex and only partially successful in correcting<br />

the exorbitism, because it carried the osteotomy<br />

in front of the orbital rim, lacrimal sac, and medial<br />

canthal ligament. In 1962 Converse and Smith 366<br />

described an operation to correct hypertelorism based<br />

on their experience with malunited nasoorbital fractures<br />

with telecanthus.<br />

Paul Tessier is regarded by most clinicians as the<br />

father of craniofacial surgery. After years of careful<br />

study with many of the most innovative maxillofacial<br />

surgeons of his time and numerous hours spent in<br />

the laboratory doing cadaver dissections, Tessier proposed<br />

a novel method of facial osteotomies and<br />

wholesale mobilization of bone for operating on<br />

patients with deformities of the craniofacial skeleton.<br />

Tessier pioneered the intracranial approach for the<br />

correction of hypertelorism, worked closely with<br />

neurosurgeons to resect difficult craniofacial tumors,<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

was first to manipulate cranial bones in the treatment<br />

of patients with craniosynostoses, and performed<br />

successful Le Fort III operations in difficult<br />

cases of Apert and Crouzon syndrome. In 1967<br />

Tessier 367 presented this work at the Fourth International<br />

Congress of <strong>Plastic</strong> and Reconstructive <strong>Surgery</strong><br />

in Rome, and the new field of craniofacial surgery<br />

was launched.<br />

Two principles fundamental to the practice of<br />

craniomaxillofacial surgery emerged from his discussion:<br />

(1) large segments of the facial and cranial skeleton<br />

can be completely denuded of their blood supply,<br />

repositioned, and yet survive completely; and<br />

(2) the eyes can be translocated horizontally or vertically<br />

over a considerable distance without impairing<br />

the vision.<br />

Tessier’s results in the late ’60s and early ’70s<br />

proved conclusively that most skeletal deformities of<br />

the face and calvarium can be corrected or at least<br />

significantly improved by appropriate surgical<br />

maneuvers. The importance of his work to the<br />

intracranial correction of hypertelorism 368–370 cannot<br />

be overemphasized. Along with Converse’s onestage<br />

procedure, it is the backbone of present-day<br />

techniques for hypertelorism correction.<br />

Basing his approach on Le Fort’s anatomic research<br />

with cadaver skulls, 371 Tessier developed the Le Fort<br />

III osteotomy for facial advancement and presented<br />

his results in 1971. His experience encompassed<br />

151 patients representing almost 500 individual malformations<br />

of the cranial and facial region. Tessier’s<br />

contributions to craniofacial surgery were reviewed<br />

in Rome in 1982 on the occasion of the 15th anniversary<br />

of Tessier’s original presentation. 372<br />

The advent of plate-and-screw fixation after<br />

maxillary and mandibular osteotomies has been<br />

of tremendous benefit to the management of congenital<br />

as well as traumatic cases of craniomaxillofacial<br />

deformity. Rigid fixation has dramatically<br />

enhanced the stability of bony fragments,<br />

improved primary bone healing, and eliminated<br />

the need for prolonged maxillomandibular fixation<br />

in the older patient. 373–378<br />

The fate of microfixation hardware during rapid<br />

calvarial growth in infants and young children has<br />

been questioned. Munro and colleagues 379 acknowledge<br />

intracranial “migration” of microplates and<br />

screws. Goldberg and colleagues 380 report cases of<br />

children undergoing revisional surgery who were<br />

found to have microscrews and plates lying beneath<br />

33


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

the inner calvarial lamina. Subsequently Goldberg<br />

381 sought to clarify the incidence of marker<br />

plate translocation and tried to identify potential<br />

clinical implications. In a retrospective review of<br />

27 pediatric patients, CT imaging demonstrated<br />

internalization of marker fixation in 14 children.<br />

The younger the patient at the time of surgery, the<br />

higher the likelihood of translocation. Patients<br />

with syndromic craniosynostosis seemed to have a<br />

greater risk of translocation than those with isolated<br />

synostosis. Longer plates had a higher propensity<br />

for movement than shorter plates. Despite<br />

these observations, the authors note no complications<br />

related to the translocation of marker plates.<br />

Papay et al 382 state that plating osteosynthesis provides<br />

a three-dimensional, stable fixation of the cranial<br />

skeleton and anatomical recontouring of the<br />

cranial vault. In their series of 20 patients who had<br />

secondary cranial remodeling within 2 years of the<br />

first surgery, “false migration” of microplates was<br />

noted in 7. Neither the sharp edges of the screws<br />

nor the ends of the stainless steel wires pierced the<br />

dura mater in any patient. Despite the lack of neurological<br />

sequelae from these fixation systems, it is<br />

their opinion that marker plate fixation for cranial<br />

vault reconstruction should be limited to those regions<br />

where additional 3D structural support is essential.<br />

Berryhill et al 383 reviewed the fate of rigid plate<br />

fixation in 96 children. There were 375 titanium<br />

plates and 1944 screws placed among the patients in<br />

the series. They found 5 cases of delayed growth,<br />

one instance of restricted growth, 9 palpable plates<br />

causing pain, 3 fluid accumulations over plates, and<br />

2 cases of meningitis. Plates were removed in 8<br />

patients. The overall complication rate was 23%.<br />

A series by Orringer reported 55 instances of plate<br />

removal. 384 The most common reason was palpable<br />

or prominent hardware (34.5%). Other reasons<br />

included loose plates (25.5%), infection (23.6%), and<br />

exposure of hardware (20%).<br />

Resorbable plating systems have enjoyed wide<br />

acceptance in craniofacial surgery. The Lactosorb<br />

(Poly-Medics, Warsaw IN) plating material is a<br />

copolymer of polylactic and polyglycolic acid.<br />

Polyglycolic acid biodegrades more rapidly than<br />

polylactic acid, and thus these combination plates<br />

resorb faster than plates made entirely of polylactic<br />

acid. 385<br />

Eppley and Sadove 386 reviewed the effects of biodegradable<br />

plates on 10 juvenile rabbits. The fixa-<br />

34<br />

tion was placed across the left coronal suture. Six<br />

months after implantation, the authors noted symmetrical<br />

frontal bone development, unaffected<br />

growth across the coronal suture, and a histologically<br />

normal underlying suture. Apparently the fixation<br />

plate stretches to accommodate growth along the<br />

underlying suture. The change in shape of the<br />

resorbable device seems to be more important than<br />

complete degradation of the material in rapidly growing<br />

bone sites.<br />

Subsequently Eppley and Sadove 387 analyzed<br />

their results with resorbable coupling fixation in<br />

20 infants with calvarial deformities. Thin, straight,<br />

resorbable plates were used for skeletal fixation<br />

after osteotomies and repositioning. A total of<br />

231 fixation devices were implanted without complications<br />

and remained trouble-free after 12 postoperative<br />

months. Wiltfang et al 388 noted that the<br />

resorbable plates were prone to intraosseous<br />

migration (as are titanium plates), but their resorption<br />

over 12–18 months obviates this problem.<br />

Multiple other clinical series with long follow-up<br />

show the Lactosorb plating system to be associated<br />

with minimal or no complications. 389–391<br />

Other areas of significant advance in the field of<br />

craniofacial surgery include radiographic improvements<br />

such as MRI scans and 3D CT scans, 392 distraction<br />

osteogenesis, increased use of microvascular<br />

techniques and tissue expansion, and better understanding<br />

of alloplastic materials.<br />

SURGERY ON THE CRANIAL VAULT AND FOREHEAD<br />

Early Treatment<br />

The rationale for early treatment in craniosynostosis<br />

is to minimize the extent of primary and secondary<br />

calvarial deformity, to allow cranial expansion<br />

and prevent the sequelae of raised intracranial<br />

pressure, and to minimize operative morbidity.<br />

The history of neurosurgical craniectomy for the<br />

correction of craniosynostosis can be divided into<br />

two basic operative approaches. In the past the<br />

standard treatment was a linear craniectomy along<br />

the course of the stenosed suture line. This treatment<br />

was associated with a high rate of recurrence<br />

of craniosynostosis, and attempts at preventing relapse<br />

by the use of polyethylene film placed around the<br />

bone edges met with little success. 393 More recently<br />

the trend has been towards a more active ana-


tomical correction of the deformitiy at the time of<br />

surgery, with less reliance on passive correction<br />

postoperatively, although many authors still advocate<br />

vertex craniectomy for young infants with sagittal<br />

synostosis. 394<br />

In 1971 Tessier 275 mobilized the lower part of the<br />

frontal bone in one piece while performing maxillary<br />

advancement in adult patients with Crouzon or Apert<br />

syndromes. Hoffmann and Mohr 395 adapted Tessier’s<br />

supraorbital advancement techniques to infants, performing<br />

a unilateral coronal supraorbital advancement.<br />

Marchac 396 later devised a technique that involved<br />

(1) rocking and advancement of the supraorbital bar<br />

with a lateral temporal spur in the form of a Z-plasty<br />

for stability and retention—the floating forehead—<br />

and (2) mobilization and rearrangement of the anterior<br />

cranial vault by means of bone grafts. Marchac<br />

originally used the floating forehead technique with<br />

frontocranial remodeling in the treatment of<br />

brachycephaly, Crouzon or Apert syndromes, and<br />

oxycephaly. 397 Marchac had originally hoped that<br />

the floating forehead technique would allow normalization<br />

of midfacial growth, but unfortunately this<br />

did not materialize.<br />

Other techniques utilize the mathematics of projection,<br />

geometry, and stress relief to produce an<br />

appropriate forehead contour. 398 Other methods<br />

treat the forehead as a totally misshapen unit that<br />

must be adjusted in all three dimensions with multiple<br />

osteotomies and cranial bone grafts to recreate<br />

the forehead/bandeau unit in jigsaw-puzzle fashion<br />

(Fig 22). 399<br />

Munro and coworkers 400 advocate 180° reversal<br />

of the entire upper cranium, while Jackson, Hide,<br />

and Barker 401 prefer frontal transposition cranioplasty<br />

for correction of frontal contour. Ortiz Monasterio<br />

and colleagues 402 combine frontal mobilization with<br />

a one-piece orbitofacial advancement in Crouzon<br />

syndrome.<br />

Operations for frontocranial remodeling have<br />

facilitated the approach to cranial as well as facial<br />

deformities and have considerably improved the overall<br />

surgical results. The vast number and variety of<br />

methods to reshape the orbital rim and cranial vault<br />

are proof that no technique is ideal in all cases. The<br />

basic concept should be to reshape all bone that is<br />

abnormal and to rearrange bone to arrive at the<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 22. Bilateral coronal synostosis and its surgical correction. A,<br />

site of anterior cranial vault and three-quarter orbital osteotomies.<br />

B, after osteotomies and reshaping and fixation of the<br />

cranioorbital region. (Reprinted with permission from Posnick<br />

JC: Craniosynostosis: Surgical Management in Infancy. In: Bell<br />

WH (ed), Orthognathic and Reconstructive <strong>Surgery</strong>. Philadelphia,<br />

WB Saunders, 1992; vol 3, p 1859.)<br />

most aesthetic contour, and the degree to which<br />

each of these maneuvers is applied differs among<br />

patients. For example, at times it may be preferable<br />

to shape the forehead by recontouring the existing<br />

frontal bone, while at other times this goal is best<br />

accomplished by replacing the frontal bone with<br />

parietal bone. Early frontal remodeling is facilitated<br />

by easy malleability of bone; rapid reossification;<br />

outward push by the growing brain; and the beneficial<br />

effect on adjacent structures of releasing the<br />

stenosed areas. 136,199 On the other hand, the earlier<br />

35


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

in infancy the cranium is reshaped, the more it can<br />

be potentially affected by growth. 403,404<br />

McCarthy et al 405 reported an adverse effect on<br />

frontal sinus development and forehead aesthetics<br />

of early frontoorbital advancement. Of 8 patients<br />

who underwent bilateral frontoorbital advancement<br />

in infancy who had been followed for at least 10½<br />

years, only one developed a frontal sinus and all had<br />

a flattened brow. Of 3 patients with unilateral<br />

advancement, one developed a frontal sinus on the<br />

operated side and the others had an obvious deformity<br />

due to discrepancy in brow projection between<br />

the operated and nonoperated sides. The authors 405<br />

advocate bilateral frontoorbital advancement for correction<br />

of plagiocephaly because of the possible asymmetry<br />

that may result from unilateral frontal sinus<br />

development.<br />

In contrast, Bartlett, Whitaker, and Marchac 406<br />

evaluated a study population of 48 children operated<br />

for plagiocephaly in infancy using both bilateral<br />

and unilateral approaches. After following the patients<br />

for a minimum of 3 years, the authors concluded<br />

that either unilateral or bilateral frontoorbital<br />

advancement produced good results, and there was<br />

no advantage to using one technique over the other.<br />

The study obviously did not follow all these infants to<br />

the time of frontal sinus development.<br />

David and Sheen 407 analyzed their results in the<br />

surgical treatment of 39 patients with Crouzon syndrome.<br />

They found that early frontoorbital advancement<br />

was universally successful in relieving elevated<br />

intracranial pressure and reducing ocular proptosis<br />

but did not prevent midfacial hypoplasia or provide<br />

definitive cosmetic correction of the deformity.<br />

Although frontofacial advancement in adults gave<br />

good long-term results, it was attended by more complications<br />

than other corrective procedures.<br />

Marchac and Renier 403,404,408 note normal facial<br />

growth after early craniofacial surgery in many<br />

nonsyndromic patients. McCarthy, 409 on the other<br />

hand, reports premature refusion of the sutures or<br />

development of turricephaly and other calvarial contour<br />

irregularities in 50 children who had early surgery<br />

for craniofacial synostosis. The mild cases<br />

received extensive strip craniectomy and the more<br />

severe or syndromic patients had strip craniectomy<br />

with frontal bone advancement. A mean 8 years<br />

after craniofacial remodeling, 20% had to be reoperated<br />

because of recurrent deformity. The authors<br />

36<br />

conclude that early surgery did not result in satisfactory<br />

occlusal relationships or midfacial form.<br />

Dufresne and associates 410 used 3D CT analysis to<br />

calculate the intracranial/ventricular increases in volume<br />

that were produced by craniofacial surgical procedures.<br />

Marsh 411 studied with 3D CT imaging the<br />

endo- and exocranial bases of patients who had early<br />

corrective surgery for craniosynostosis. He concluded<br />

that surgery during infancy induces normalization of<br />

endocranial symmetry over the first postoperative<br />

year in patients with nonsyndromic solitary and<br />

bicoronal synostosis, but not in those with multiple<br />

synostoses. Other analyses of long-term results 412<br />

find less improvement in endocranial base symmetry<br />

or normalization of intracranial growth after early<br />

surgery.<br />

The optimal age of the patient at surgery remains<br />

controversial. Some authors strongly favor early treatment<br />

of craniosynostosis during infancy (which is variously<br />

interpreted as the period between the first week<br />

of life up to 4–6mo 393 ) to allow brain growth to<br />

remodel the calvarium. 413<br />

Ocampo and Persing 237 feel that the quality of the<br />

result is inversely proportional to age of the child at<br />

operation. Most craniofacial surgeons would agree<br />

that early surgery is indicated in the event of multiple<br />

suture synostosis 414 and in sagittal synostosis when an<br />

extended vertex craniectomy is performed. Certainly<br />

surgery should be done as early as possible for the<br />

severe craniosynostosis syndromes such as kleeblattschadel.<br />

396,404 Depending on the number and location<br />

of prematurely fused sutures, therefore, early<br />

cranial vault decompression may be indicated to prevent<br />

restrictions on the developing brain. 403,411,415 It<br />

is impossible to know which infants will require early<br />

release to prevent neural complications.<br />

By age 18 months, children have attained<br />

approximately 75% of their brain growth 136,411 (Table<br />

7). Reossification is more extensive at a younger<br />

age 416 and can be reliable up until age 2.<br />

When single sutures are involved, many craniofacial<br />

surgeons delay surgery until the child is 6–12<br />

months, at which time the risk of intracranial hypertension<br />

is thought to be low. 404,411 Wall 417 notes a<br />

higher reoperation rate when patients are operated<br />

on at an early age (3mo) than if frontoorbital<br />

advancement is delayed (6–12mo). Because the<br />

changes brought about by surgery will be diluted by<br />

postoperative growth, others 415 wait even longer in<br />

cases of Apert syndrome and the more severe sym-


TABLE 7<br />

Brain Growth During the First 20 Years of Life<br />

(Data from Blinkov SM, Glezer II: The Human Brain in Figures<br />

and Tables: A Quantitative Handbook. New York, Plenum<br />

Press, 1968. Reprinted with permission from Marchac D, Renier<br />

D: <strong>Craniofacial</strong> <strong>Surgery</strong> for Craniosynostosis. Boston, Little<br />

Brown, 1982, p 36.)<br />

metrical deformities to lessen the need for major<br />

revisions at a later date. 404<br />

Certainly unilateral cases can be expected to<br />

achieve better results than bilateral cases. McCarthy<br />

demonstrated good to excellent results in 86% of<br />

patients undergoing unilateral procedures and in 49–<br />

70% of bilateral procedures. 418,419<br />

SURGERY FOR COMBINED CRANIOSYNOSTOSIS AND<br />

HYPOPLASIA OF THE MIDFACE<br />

There have been few changes in the treatment of<br />

Crouzon disease since Tessier first described his classic<br />

facial osteotomy technique. 275,288,367,420,421 Modifications<br />

have progressed from the Tessier I—a<br />

subcranial Le Fort III—in 1958, through the more<br />

radical Tessier VIII of 1976, which split and bent a<br />

frontofacial monobloc segment, derotated the orbits,<br />

and brought the maxilla forward. In Tessier’s hands<br />

this extensive procedure yielded nearly normal results.<br />

In 1976 and again in 1979, Van der Meulen 422,423<br />

described a similar operation that involves simultaneous<br />

total osteotomy of the two halves of the face<br />

and correction of the orbitofrontal and maxillary<br />

deformities. The coronal approach to osteotomy of<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

the pterygomaxillary fissure obviates intraoral surgery<br />

with its potential contamination.<br />

Ortiz Monasterio and associates 402 first advocated<br />

monobloc advancement of the entire forehead and<br />

face (Fig 23) in 1977. Anderl and coworkers 424 proposed<br />

a modification of the frontoorbital bar with<br />

preservation of an intact anterior cranial base to separate<br />

the cranial and nasal spaces when indicated for<br />

relief of respiratory or orbital distress accompanying<br />

severe craniosynostosis. Although many surgeons<br />

believe that monobloc advancements are hazardous<br />

because of the high number of associated infectious<br />

sequelae, the authors feel their technique is safe even<br />

at a young age.<br />

Fig 23. Cranial vault reshaping for correction of Crouzon<br />

syndrome with bilateral coronal synostosis. The procedure<br />

involves osteotomies of the anterior cranial vault, monobloc, Le<br />

Fort I, and chin. (Reprinted with permission from Posnick JC:<br />

Craniosynostosis: Surgical Management of the Midface Deformity.<br />

In: Bell WH (ed), Orthognathic and Reconstructive<br />

<strong>Surgery</strong>. Philadelphia, WB Saunders, 1992; vol 3, p 1888.)<br />

Wolfe 425 reviewed his experience with 32 patients<br />

who underwent transcranial monobloc frontofacial<br />

advancement +/– simultaneous facial bipartition.<br />

The author cautions that the procedure “carries with<br />

it substantial risks, [but] with careful consideration of<br />

airway control, the anterior cranial base dura, and<br />

retrofrontal dead space, the procedure is recommended<br />

for carefully selected patients.” Wolfe lists<br />

his indications and contraindications for the procedure<br />

in various age groups.<br />

Treatment for combined deformities from craniosynostosis<br />

of the anterior cranial vault and<br />

37


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

midfacial retrusion typically involves two operative<br />

stages, initially advancing the forehead and subsequently<br />

the face. This results in fewer infections<br />

than monobloc advancement. 426 Marchac and<br />

Renier 397 advocate Tessier’s frontal horizontal<br />

advancement with lateral fixation by tongue-ingroove<br />

for intracranial forehead correction. The<br />

resultant exaggeration in facial retrusion is corrected<br />

at a later date. 397,404<br />

Fearon and Whitaker 426 reviewed their 15-year,<br />

29-patient experience with midfacial and<br />

frontofacial advancement for craniosynostosis. Of<br />

the 30 procedures performed, 20 were Le Fort IIIs<br />

and 10 were monobloc advancements. All major<br />

infections occurred in the monobloc group. The<br />

aesthetic results were the same regardless of the<br />

type of facial advancement performed. Given the<br />

high infection rate associated with monobloc<br />

advancement and the similar outcomes, the<br />

authors believe the surgical correction of craniosynostosis<br />

should be staged.<br />

SURGERY ON THE ORBITS<br />

Surgical techniques for the correction of exorbitism,<br />

ocular dystopia, enophthalmos and hypertelorism are<br />

based on principles of craniofacial surgery.<br />

Dystopia<br />

Ocular dystopia can be congenital or traumatic<br />

and is corrected by unilateral orbital repositioning. 427<br />

Edgerton and Jane428 review various eye-leveling techniques,<br />

ranging from simple bone grafts to complex<br />

C-type osteotomies429 and Tessier’s marginal<br />

osteotomy, which removes the orbital rim, levels it,<br />

and repositions it.<br />

The surgical treatment of ocular dystopia varies<br />

according to the age of the patient and the cause<br />

of the orbital anomaly. In young patients who are<br />

actively growing, it is preferable to use the<br />

frontoorbital bandeau technique so as not to damage<br />

the dental buds. In adult patients, Tessier’s430 orbital quadrant technique is used. At birth the<br />

orbital volume is about 70% of its adult volume.<br />

At 18 months, it is about 75–80%, and at 3 years<br />

of age, it is about 90%. The orbital volume is<br />

completely developed by age 10 to 11.<br />

38<br />

Hypertelorism<br />

The severity of hypertelorism is rated according to<br />

the interorbital distance (IOD), 370 as follows:<br />

1° — 30–35mm IOD<br />

2° — 35–39mm IOD with normal orientation<br />

and shape of the orbits<br />

3° — 40+mm IOD with defects in the<br />

cribriform plate, orbital region, and<br />

lateral canthus<br />

The normal adult IOD is 25mm for women and<br />

28mm for men. The usual growth progression 431 is<br />

as follows:<br />

age 1 year 18.5mm<br />

3 years 21mm<br />

7 years 23mm<br />

12 years 26mm<br />

The interpupillary distance (IPD) is not a true indication<br />

of hypertelorism because abnormal rotation or<br />

divergent strabismus of the eyes commonly accompanies<br />

the syndromic craniofacial anomalies. Similarly, the<br />

intercanthal distance or ICD can be skewed by excessive<br />

soft tissue bulk or traumatic telecanthus.<br />

Evereklioglu et al 432 propose a clinically applicable<br />

and practical definition of hypertelorism as an<br />

increase in the interpupillary distance in the absence<br />

of strabismus (Fig 24). An isolated measurement of<br />

hypertelorism is not clinically applicable unless it is<br />

noted in the context of overall head size. The authors<br />

propose an interpupillary index which they believe is<br />

a more accurate projection of hypo- and hypertelorism<br />

related to head size. The index is calculated<br />

by the formula<br />

The authors also provide normative values in healthy<br />

children.<br />

Hypertelorism is etiologically heterogeneous.<br />

Nontraumatic hypertelorism is always secondary to<br />

another deformity, either craniosynostosis or a median<br />

or paramedian facial cleft. Munro 427 notes four types<br />

of ethmoid and medial orbital wall deformity in<br />

hypertelorism (Fig 25). Types C and D are the rarest<br />

and most difficult to correct.<br />

Correction of hypertelorism can be subcranial or<br />

intracranial. The subcranial approach offers two<br />

alternatives: medial orbital wall migration or U-shaped


Fig 24. Interocular distance measured from inner canthus to<br />

inner canthus. A, normal. B, primary telecanthus. C, true ocular<br />

hypertelorism. D, ocular hypertelorism with secondary<br />

telecanthus. (Reprinted with permission from Cohen MM Jr,<br />

Richieri-Costa A, Guion-Almeida ML, Saavedra D: Hypertelorism:<br />

interorbital growth, measurements, and pathogenetic considerations.<br />

Int J Oral Maxillofac Surg 24:387, 1995.)<br />

osteotomy mobilizing the lateral, inferior, and medial<br />

orbital walls as a single bloc. The maximum correction<br />

that is possible with medial orbital wall mobilization<br />

is 10mm; with a U-shaped osteotomy, 15mm.<br />

The intracranial approach as described by<br />

Tessier 368,369 with Converse’s modification 433 is preferred<br />

in all severe cases of hypertelorism or when<br />

the cribriform plate lies below the level of the<br />

frontonasal suture (Fig 26). The entire orbit is mobilized<br />

as a box, providing total control of the intracranial<br />

contents.<br />

McCarthy and colleagues 434 report correction of<br />

orbital hypertelorism on 20 children age


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 26. Above, one-stage medial orbital osteotomy with preservation<br />

of the cribriform plate. Below, subtotal orbital osteotomy<br />

with preservation of the cribriform plate. (Reprinted with permission<br />

from Converse JM, Wood-Smith D: An atlas and classification<br />

of mid-facial and craniofacial osteotomies. In: Transactions<br />

of the Fifth International Congress of <strong>Plastic</strong> and<br />

Reconstructive <strong>Surgery</strong>. Melbourne, Feb 1971, p 931.)<br />

Anophthalmia and Microphthalmia<br />

Anophthalmos and microphthalmos represent failure<br />

of optic development. Absence or diminutive<br />

size of the eyes fails to stimulate orbital growth and<br />

results in orbital hypoplasia. Kennedy437 elicited<br />

hypoplasia of the orbit by ocular enucleation during<br />

embryogenesis in cats.<br />

The classic treatment for orbital hypoplasia secondary<br />

to anophthalmia or microphthalmos has been<br />

the insertion of progressively larger silicone orbital<br />

implants covered by scleral shells. This treatment<br />

has met with only moderate success due to difficulty<br />

in maintaining the implants and the need for frequent<br />

readjustments.<br />

Tessier438 enlarged the orbit by a lateral<br />

osteotomy that allowed expansion of the orbital<br />

volume inferiorly and laterally. Marchac439 later<br />

added a supraorbital craniectomy and frontal craniotomy<br />

that relocated the orbital roof and allowed<br />

expansion of the orbital volume in three directions.<br />

Elisevich and colleagues440 subsequently<br />

40<br />

described a technique for 3D expansion of the<br />

orbit without craniotomy using a single burr-hole<br />

craniectomy of the frontosphenoid bone.<br />

Working from the basis that orbital hypoplasia is<br />

the consequence of an inadequate stimulating matrix,<br />

Lo et al 441 demonstrated the effectiveness of subperiosteal<br />

tissue expanders in the bony orbit to substitute<br />

for the missing stimulus. They enucleated one eye in<br />

12 6-week-old kittens and placed subperiosteal tissue<br />

expanders in half of the orbits; the other 6 served<br />

as controls. All the orbits in which tissue expanders<br />

were placed demonstrated equal or greater orbital<br />

volumetric measurements than normal orbits, compared<br />

to enucleated orbits that did not receive tissue<br />

expanders, whose measurements were significantly<br />

lower than the normal.<br />

Rodallec and coworkers 442 report the use of silicone<br />

expanders in 17 patients. The expanders were<br />

placed within the intramuscular cone to expand not<br />

only the bony orbit but also the soft tissues.<br />

SURGERY ON THE MAXILLA<br />

Le Fort I Osteotomy<br />

The Le Fort I osteotomy is a horizontal osteotomy<br />

of the upper jaw at the level of the (Le Fort I) fracture<br />

line. It was initially described by von Langenbeck in<br />

1859 and used widely in the second half of the 19th<br />

century to temporarily mobilize the maxilla for better<br />

access to tumors and polyps. Not until the 1960s,<br />

however, did mobilization of the maxilla become<br />

generally accepted, due in large measure to the precise<br />

operative technique and superb results reported<br />

by Obwegeser, 373 who demonstrated that it was possible<br />

to fully mobilize the maxillary segments and<br />

bring them into the desired position without softtissue<br />

resistance for primary stabilization. 443<br />

Obwegeser also described a higher maxillary<br />

osteotomy (Le Fort I½) for use in patients with pronounced<br />

midfacial recession to advance the retruded<br />

portion of the maxilla lateral to the piriform aperture.<br />

Other modifications of the Le Fort I osteotomy<br />

have been described, including a self-stabilizing<br />

osteotomy suggested by Munro—a “self-retained Le<br />

Fort I”. 444<br />

Many authors have confirmed tooth viability subsequent<br />

to Le Fort I osteotomy, with reinnervation of<br />

the pulp over a period of several months. 445,446


Le Fort II Osteotomy<br />

The Le Fort II osteotomy was popularized by<br />

Henderson and Jackson447 in cleft patients with associated<br />

paranasal and nasal shortening. Several variations<br />

have been described, including the tripartite<br />

osteotomy of Converse. 448<br />

Le Fort III Osteotomy<br />

The mainstay of total maxillary advancement is<br />

the Le Fort III osteotomy. 421 The procedure can be<br />

combined with other osteotomies, specifically a Le<br />

Fort I, for definitive treatment of severe midfacial<br />

retrusion. 288 Interpositional bone grafts and a<br />

maxillomandibular fixation appliance373–377 are useful<br />

adjuncts. If decreasing the interorbital distance or<br />

widening of the palate is necessary, a facial bipartition<br />

is indicated (Fig 27).<br />

A long-term study by Bachmayer et al449 of maxillary<br />

growth after Le Fort III osteotomy in syndromic<br />

craniosynostosis showed minimal (mean 5%) vertical<br />

relapse with rigid stabilization of the midface, but<br />

severe anteroposterior growth retardation . Although<br />

the average forward movement of the maxilla at the<br />

time of surgery was 12.4mm, horizontal maxillary<br />

growth was minimal after surgery, and subsequent<br />

maxillary advancement was invariably required at<br />

the completion of growth.<br />

In a retrospective study of patients who underwent<br />

Le Fort III advancement for craniofacial dysostosis,<br />

Kaban and colleagues450 note early skeletal<br />

relapse in the sagittal plane in approximately 33% of<br />

patients. The magnitude of relapse was greater in<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

patients with Apert syndrome than in those with<br />

Crouzon disease. The small amount of relapse during<br />

the first year was followed by progressive downward<br />

and forward movement of the midface until, 2<br />

years after surgery, the midface was either anterior<br />

or inferior to the immediate postoperative position<br />

in 15 of 19 patients. Three patients required an<br />

additional Le Fort I osteotomy to correct Class III<br />

malocclusion.<br />

A prospective analysis of 12 “growing” patients by<br />

McCarthy and colleagues 451 disclosed a remarkable<br />

degree of postoperative skeletal stability. They followed<br />

this study with a clinical and cephalometric<br />

longitudinal analysis of 12 children with craniofacial<br />

synostosis syndromes who underwent Le Fort III<br />

advancement at age


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

morbidity and improve results include hypotensive<br />

anesthesia, shorter operative time, rigid stabilization<br />

of the mobilized bones at the end of the operation,<br />

fewer incisions, extensive antibiotic therapy, and<br />

greater surgeon’s experience.<br />

Munro and Sabatier 454 analyzed the results of<br />

craniofacial surgery in 1092 patients representing<br />

2019 procedures. Complications of surgery<br />

accounted for 0.18% of deaths. Major complications<br />

occurred in 14.3% of patients but not all had<br />

permanent sequelae. Infection was the greatest<br />

problem, occurring in 5.3%. As the surgeons’<br />

experience increased, the complication rate<br />

decreased.<br />

David 455 reviewed his 10-year experience with<br />

craniofacial surgery in Australia. The overall infection<br />

rate in 170 patients was 6.5%, with a marked<br />

age differential: 23.5% in adults and 2.2% in children.<br />

The highest infection rates were seen in<br />

patients who required tracheostomy and had longer<br />

operative times, and the most common pathogen<br />

was Pseudomonas. His recommendations to prevent<br />

infectious complications include operative<br />

intervention at an early age, short preoperative hospital<br />

stay, antibiotic prophylaxis to include gramnegative<br />

cover, isolating the dead space, and strict<br />

tracheostomy care.<br />

Poole 456 analyzed the postoperative course of 200<br />

consecutive patients treated at the Oxford <strong>Craniofacial</strong><br />

Unit. Their overall complication rate was 22%,<br />

including 1% mortality and 1% infections. In this<br />

series as well as in those mentioned above, the frequency<br />

of infection was greater in adults than in<br />

children and intracranial procedures carried a higher<br />

risk than extracranial procedures.<br />

Whitaker and colleagues 415 retrospectively analyzed<br />

the records of 164 patients with craniosynostosis<br />

operated on over a 12-year period. The<br />

authors state that excellent results can be expected<br />

in the asymmetrical deformities treated in infancy<br />

by a unilateral approach. Similarly, early surgery in<br />

the form of bilateral orbital advancement gives satisfactory<br />

results in most patients with mild symmetrical<br />

deformities. In contrast, cases of severe bilateral<br />

deformity or Apert syndrome often required a second,<br />

frequently extensive, operation. There were<br />

no deaths, blindness, or brain dysfunctions as a<br />

result of surgery.<br />

42<br />

DISTRACTION OSTEOGENESIS<br />

History<br />

Distraction osteogenesis (DO) is the process by<br />

which new bone is generated in the gap between<br />

two bone segments in response to the application of<br />

graduated tensile stress across the gap. DO was first<br />

described by Codivilla in 1905. 457 Beginning in the<br />

early 1950s, Ilizarov458 introduced the concept of<br />

distraction osteogenesis for acquired and congenital<br />

deformities of enchondral bone. 459 In 1973 Snyder460 reported distraction of the mandible in dogs, and 20<br />

years later McCarthy et al57 described lengthening of<br />

the mandible in 4 patients with hemifacial microsomia<br />

by application of DO.<br />

In 1998 Molina and Ortiz-Monasterio published<br />

their series of mandibular distraction in 274 patients, 58<br />

and the following year McCarthy reviewed his 10year<br />

experience with distraction of the mandible. 461<br />

Habal462 reported midfacial distraction using an<br />

external distractor in 1995. That same year Polley463 described monobloc craniomaxillofacial distraction.<br />

Chin and Toth464 developed an internal device for<br />

midface advancement at the Le Fort III level in 1997.<br />

Biology of Distraction<br />

The process of distraction allows for the generation<br />

of bone—osteogenesis—as well as soft-tissue—<br />

histogenesis. In a gap between two bone segments<br />

initially a hematoma forms, which then develops into<br />

a soft callus. The application of tensile stress prevents<br />

progression to hard callus and subsequent fracture<br />

union. Instead, intramembranous bone formation<br />

occurs within the ever increasing gap. Within<br />

the bony gap there are three zones. The fibrous<br />

interzone intervenes between the two primary mineralizing<br />

fronts adjacent to the osteotomy. At cessation<br />

of distraction the mineralizing fronts meet and<br />

unite to create union during the consolidation phase.<br />

The resultant bony union is dependent on the latency,<br />

rate, rhythm, and consolidation period of the distraction<br />

process.<br />

Latency. Latency is the time period after<br />

osteotomy/corticotomy and before commencing distraction.<br />

In an ovine model Tavakoli et al 465 found<br />

no difference in mechanical strength or bone density<br />

of the callus when the latency period varied between


0 and 7 days. This would suggest that the traditional<br />

practice of allowing a nontreatment interval may be<br />

unnecessary and may only prolong treatment time.<br />

Cedars 466 uses no latency period during Le Fort III<br />

distraction.<br />

Rate. Rate is the number of millimeters the bone<br />

gap is widened each day. Farhadieh and<br />

Gianoutsos 467 used a sheep model comparing distraction<br />

rates of 1mm, 2mm, 3mm, and 4mm per<br />

day. They then measured biomechanical properties,<br />

mineralization, and histology of the regenerate.<br />

They found a distraction rate of 1mm per day<br />

was superior to all others. A rate of 1mm/d also<br />

gave the most uniform bone formation radiologically<br />

and histologically in a porcine model studied<br />

by Troulis. 468 When distracting at the Le Fort III<br />

level, Cedars et al 466 uses a torque wrench to determine<br />

the rate of distraction, believing this reflects<br />

the tolerance of the soft-tissue envelope and is more<br />

appropriate than an arbitrary measurement.<br />

Rhythm. Rhythm is the number of times daily the<br />

distractor is activated. Rhythm varies from once per<br />

day to 4 times per day.<br />

Consolidation. A long consolidation period will<br />

lead to weakening of the regenerate as a result of<br />

disuse atrophy, 469 whereas too short a consolidation<br />

phase will lead to fibrous nonunion, buckling or fracture<br />

of the regenerate. 470 Rachmiel et al 471 showed<br />

24% mineralization after 3 weeks of consolidation<br />

and 77.8% by the end of 6 weeks.<br />

To date little clinical research has been done into<br />

the appropriate length of the consolidation period.<br />

Smith et al 472 used computed tomography to suggest<br />

6 weeks was the minimum amount of time the<br />

regenerate should be allowed to mineralize prior to<br />

device removal. Felemovicius 473 used bone scintigraphy<br />

with technetium-99 diphosphonate to assess the<br />

duration of the consolidation period. They found<br />

consolidation underway at 5 weeks in infants under<br />

12 months old, while in older children it was ongoing<br />

at 10 weeks and in adolescents and adults, at 10-<br />

14 weeks.<br />

Distraction Treatment Planning<br />

Compared with the relatively simple distraction of<br />

long bones, distraction of the mandible in the hypo-<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

plastic deformity is decidedly a challenge. A treatment<br />

plan is essential to design the 3D vectors necessary<br />

to correct mandibular deficiency. Molina 474 states<br />

that the most critical factor in mandibular DO is<br />

proper planning of the distraction vector on the<br />

cephalogram and its replication in the operating room.<br />

The hypoplastic ramus must be elongated, bringing<br />

the affected side of the jaw downward, forward, and<br />

toward the midline.<br />

Tharanon and Sinn 475 describe 3D treatment<br />

planning of mandibular hypoplasia using a panoramic,<br />

postero-anterior (PA), and lateral<br />

cephalogram. From the panoramic cephalogram,<br />

the site for the osteotomy is chosen to avoid damage<br />

to the inferior alvelolar nerve and the tooth<br />

buds. The necessary vertical lengthening is determined<br />

from the PA cephalogram, to bring the<br />

angles of the mandible onto an equal plane. From<br />

the lateral cephalogram, the required horizontal<br />

lengthening is determined based on correction of<br />

the central incisors and chin to the desired position<br />

(Fig 28). The authors advocate this method<br />

on the basis that operative time is saved, the<br />

patient’s family can be informed as to the duration<br />

of the distraction phase, and the technique is<br />

simple and inexpensive. Orientation of the vector<br />

of distraction parallel to the sagittal plane avoids<br />

lateral displacement and bending forces on the<br />

bone. 472<br />

Gateno et al 476 described a technique of 3D modeling<br />

and animation they employed in 7 patients to<br />

simulate distraction osteogenesis in virtual reality. The<br />

technique requires familiarity with advanced animation<br />

software, is time consuming, and a surgical template<br />

must be constructed and used intraoperatively<br />

to ensure correct pin placement. 477<br />

Distraction Devices<br />

The initial devices developed for distraction<br />

osteogenesis were external and unidirectional. More<br />

recent devices operate in multiple planes and multiple<br />

vectors, from Pensler’s 478 ball and socket joint<br />

to McCarthy’s 479 multiplanar distractor that works<br />

in the sagittal, vertical, and coronal planes simultaneously<br />

(Fig 29). McCarthy 479 emphasizes the following<br />

technical points in the application of<br />

multiplanar distraction:<br />

43


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 28. Mandibular distraction with an extraoral device in hemifacial microsomia. A, location of the osteotomy. B, C, pin positions. D,<br />

amount of vertical mandibular distraction on the affected side. E, overbite relationship between the maxillary and mandibular central<br />

incisors. F, center of rotation. The distal segment is moved horizontally until the overjet and chin are in the best position. The horizontal<br />

movement determines the actual distance of AP distraction. (Reprinted with permission from Tharanon W, Sinn DP: Mandibular<br />

distraction osteogenesis with multidirectional extraoral distraction device in hemifacial microsomia patients: three-dimensional<br />

treatment planning, prediction tracings, and case outcomes. J Craniofac Surg 10:202, 1999.)<br />

• linear distraction continues at all times to avoid<br />

premature consolidation<br />

• linear distraction is begun first to generate<br />

adequate bony regenerate<br />

• both limbs of the device are available for linear<br />

distraction and can be varied appropriately<br />

• linear distraction is by far the most important and<br />

often the only vector required<br />

The development of intraoral devices was spurred<br />

by a desire to minimize external scarring. The intraoral<br />

44<br />

devices can be tooth/bone borne 480 and located<br />

intraorally (Guerrero 481 reports more than 200 distractions<br />

using this type of device), or bone borne<br />

and located submucosally or buried. 482<br />

Rachmiel et al 483 compared the results of internal<br />

and external devices in mandibular DO. In a<br />

poulation of 22 patients with hemifacial microsomia,<br />

12 had DO by an external device and 10 by<br />

an intraoral device. The external device allowed<br />

greater elongation and better extraoral vector control,<br />

and conserved the gonial angle. The main<br />

disadvantages of the external device were the<br />

Fig 29. Multiplanar mandibular distraction device to achieve simultaneous yet independent linear distraction (sagittal plane), angular<br />

distraction (vertical plane), and transverse distraction (coronal plane). (Reprinted with permission from McCarthy JG, Williams JK,<br />

Grayson BH, Crombie JS: Controlled multiplanar distraction of the mandible: device development and clinical application. J Craniofac<br />

Surg 9:322, 1998.)


inconvenience to the patient and the cutaneous scars.<br />

The intraoral device was more socially acceptable<br />

and left no visible scars, but there was loss of vector<br />

control and a second procedure was required to<br />

remove the device.<br />

Polley 484 reported on an external halo-mounted<br />

device for midfacial monobloc DO in a newborn.<br />

Chin and Toth 464 and Cohen 486 developed internal<br />

devices for midface advancement (Fig 30), citing<br />

the advantage of patient convenience;<br />

Cohen’s 486,487 device was resorbable to eliminate<br />

the need for a removal procedure. Motor-driven<br />

distraction devices have also been reported. 488<br />

External devices are most commonly used in the<br />

midface. 489<br />

Fig 30. Patient with repaired cleft lip and severe midfacial<br />

hypoplasia. A, B, before distraction. Middle, patient fitted with<br />

rigid external distraction apparatus for correction of midfacial<br />

deficiency. C, D, after maxillary distraction. (Reprinted with<br />

permission from Polley JW, Figueroa AA, Kidd M: Priciples of<br />

Distraction Osteogenesis in <strong>Craniofacial</strong> <strong>Surgery</strong>. In: Lin KY,<br />

Ogle RC, Jane JA (eds), <strong>Craniofacial</strong> <strong>Surgery</strong>. Science and<br />

Surgical Technique. Philadelphia, WB Saunders, 2002; Ch 11,<br />

pp 163-171.)<br />

<strong>Craniofacial</strong> Distraction<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Mandible<br />

McCarthy’s protocol for mandibular distraction is<br />

based on patient age and type of mandibular deformity,<br />

as follows: 461<br />

Under 2 years – DO of the mandible is not indicated<br />

Age 2-6 years –<br />

Pruzansky Type I – orthognathic surgery including<br />

DO are deferred until adolescence490,491 Pruzansky Type I (severe) and Type II –<br />

DO is indicated<br />

Pruzansky Type III – Stage I: costochondral rib<br />

graft at age 3-4 yrs; if absent, the glenoid is<br />

reconstructed with a rib graft and fixed to<br />

the zygoma. Stage II: DO of the rib graft at<br />

6 months postinsertion of costochondral rib<br />

graft<br />

Age 6-teenager – primary DO is considered if no<br />

previous treatment. Secondary DO is<br />

considered in patients with significant deformity<br />

after rib grafting.<br />

Teenager – surgery is postponed until skeletal maturity<br />

is reached.<br />

Post skeletal maturation – minimal deformities are<br />

best treated with classic orthognathic surgery. 492<br />

Mandibular distraction should be considered in<br />

patients with moderate to severe deformity and<br />

bilateral disease. 493<br />

McCormick and colleagues 494,495 investigated the<br />

effect of osteodistraction on the temporomandibular<br />

joint (TMJ), first in a canine model and then in human<br />

subjects. Compression on the cartilaginous portion<br />

of the condylar head has been shown to be detrimental<br />

to subsequent TMJ morphology and function,<br />

496–498 yet in this series initial condylar flattening<br />

was transient and completely reversible. In fact, bone<br />

distraction appears to improve the temporomandibular<br />

joint. 499 When properly applied, the distraction<br />

forces pull the mandible in a downward and forward<br />

direction, leading to new bone deposition along<br />

the posterior aspect of the mandible and resorption<br />

along the anterior ramus region. 500 The overall conclusion<br />

of these studies was that distraction was beneficial<br />

to the TMJ complex.<br />

Speech before and after mandibular DO was<br />

assessed by Guyette et al, 501 who report worse<br />

articulation and nasal resonance post-distraction.<br />

These changes were temporary, however, and in<br />

45


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

time all patients returned to their pre-distraction<br />

speech levels.<br />

The actual vector of distraction achieved may vary<br />

from that planned. This may be due to technical<br />

error or to the deforming forces of the soft-tissue<br />

envelope. Maloclusion, an anterior open bite, or an<br />

unaesthetic result can be adjusted by manipulation<br />

of the callus prior to mineralization. Hoffmeister et<br />

al 502 introduced the “floating bone” concept of elastics<br />

to reshape the regenerate and improve occlusion.<br />

Kunz 503 uses elastics during and after distraction<br />

to correct minor deviations of the distraction<br />

vector, a technique he calls “a lifeboat.” Pensler 478<br />

employs direct manual reshaping of the callus to<br />

achieve the same end. Early removal of the device<br />

(at 3 weeks) and orthodontic elastics can close an<br />

open lateral bite when an undesirable vector is<br />

obtained. 483<br />

In the neonatal population, mandibular distraction<br />

has been used to help manage the airway.<br />

Denny 116 reports a series of 10 patients with mandibular<br />

hypoplasia who were successfully treated with<br />

DO. After distraction was completed, 2 of 3 patients<br />

who had tracheostomies were successfully decannulated.<br />

Denny 504 later demonstrated that mandibular<br />

advancement via distraction osteogenesis could<br />

also be used to avert tracheostomy in neonates with<br />

craniofacial anomalies that impinge on the airway.<br />

Le Fort I<br />

Before the pubertal growth spurt, DO of the mandible<br />

will allow descent of the maxilla orthodontically<br />

to a more normal position. After the pubertal growth<br />

spurt, combined DO of the maxilla and mandible<br />

will be necessary because of the cessation of maxillary<br />

growth. Molina and Ortiz Monasterio58,505 describe a technique involving Le Fort I corticotomy<br />

at the time of mandibular osteotomy and distractor<br />

placement. After a 5-day latency, intermaxillary wiring<br />

of the jaws is performed and distraction begun.<br />

Consolidation extends for 8 weeks.<br />

Molina and Ortiz Monasterio506 also report their<br />

experience with distraction of the maxilla in isolation<br />

in 36 patients, 18 with unilateral cleft lip and palate,<br />

12 with bilateral cleft lip and palate, 2 with<br />

nasomaxillary dysplasias, and 4 with mandibular<br />

prognathism. All patients were between the ages of<br />

3 and 11 years at the time of operation. The distraction<br />

technique involved a vestibular incision, subpe-<br />

46<br />

riosteal dissection of the maxillae, and incomplete<br />

osteotomy above the canines and molar roots. The<br />

osteotomy must respect the integrity of the maxillary<br />

antral mucoperiosteum and should extend into the<br />

maxillary buttress. Pterygomaxillary dysjunction and<br />

dissection along the nasal floor and base of the septum<br />

are not done. On the fifth postoperative day,<br />

the authors hook a maxillary orthopedic appliance<br />

to a facial mask with rubber bands and 950g of<br />

mechanical force is applied to each side. Each week<br />

the maxilla is advanced about 3-4mm. Once half of<br />

the total projected maxillary advancement (8-12mm)<br />

and a Class II molar relationship have been obtained,<br />

the force is decreased to one rubber band on each<br />

side to promote new bone formation at the osteotomy<br />

site and pterygomaxillary junction. The overall<br />

improvement was reported as excellent.<br />

Cohen 485 applied the MIDS internal device for Le<br />

Fort I advancement in 2 patients and reports 15mm<br />

and 17mm of advancement (Fig 31).<br />

Ko et al 507 examined the soft-tissue response to<br />

DO of the maxilla in 16 patients. They found that<br />

the concave facial profile became convex, the<br />

advancement of soft-tissue to hard-tissue ratio at A<br />

point was 0.96:1, and the soft-tissue to hard-tissue<br />

ratio for the nasal tip was 0.53:1. In a separate<br />

study, Ko et al 508 reported on the effects of maxillary<br />

DO on the speech of 22 patients. The average<br />

maxillary advancement was 8.9mm and resulted in<br />

14% worsening of hypernasality. The hypernasality<br />

was apparently related to the degree of advancement<br />

and unaffected by the presence of a pharyngeal<br />

flap. Despite worsening of nasal air escape,<br />

57% of patients had improved articulation.<br />

Le Fort III/Monobloc<br />

Midface distraction was first performed in 1993. 509<br />

In 1995 Polley484 used an external distraction device<br />

to perform a monobloc distraction in a newborn,<br />

and in 1997 the same author described applications<br />

of the external device for midface distraction in childhood<br />

and adolescence. In 1996 Chin and Toth464 reported on Le Fort III distraction with an internal<br />

buried device. Distraction advancement of up to<br />

25mm has been described. 510<br />

In younger patients who require frontoorbital<br />

advancement as well as advancement of the midface,<br />

monobloc DO is appropriate. In older patients who<br />

have undergone previous frontorbital advancement


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Fig 31. Modular internal distraction system applied in left, monobloc osteotomy and right, Le Fort III osteotomy. (Reprinted with<br />

permission from Cohen SR: <strong>Craniofacial</strong> distraction with a modular internal distraction system: evolution of design and surgical<br />

techniques. Plast Reconstr Surg 103:1592, 1999.)<br />

DO at the Le Fort III level, this may be all that is<br />

required. Both internal and external devices exist for<br />

midfacial DO. In younger patients the hypoplastic<br />

malar complex may not withstand the forces of distraction<br />

511 and an external appliance attached to the<br />

maxillary dental arch may be more appropriate.<br />

Cedars et al 466 reported a series of 14 patients<br />

who underwent Le Fort III DO with the internal<br />

device developed by Chin and Toth. 464 Following<br />

osteotomy and application of the device, the distraction<br />

pins exit percutaneously through the cheek, and<br />

a torque wrench is used to begin distraction intraoperatively.<br />

Distraction is continued immediately postoperatively<br />

using the torque wrench twice a day to a<br />

load of 14–18N-cm. DO is continued until the<br />

porion–orbitale distance is achieved as planned<br />

cephalometrically. The average distraction was 18mm<br />

(range 12.5–27mm; 11mm achieved intraoperatively).<br />

Cephalometric follow-up for more than 1<br />

year in 6 patients showed excellent stability of the<br />

advancement at the occlusal level, with some relapse<br />

at the level of the orbitale. All patients had improvement<br />

in symptoms of sleep apnea. One of 2 patients<br />

with tracheostomy was decannulated. The authors<br />

now concentrate the distraction process on achieving<br />

superior maxillary position, reserving height<br />

increase for Le Fort I osteotomy to be performed<br />

subsequently.<br />

Fearon 512 compares the results of Le Fort III<br />

advancement in a pediatric series of 22 patients. Ten<br />

patients underwent standard Le Fort III advancement<br />

and 12 underwent advancement by DO (2<br />

internal buried device, 10 external halo device). In<br />

the standard Le Fort III group, the average advancement<br />

was 6mm; in the distracted group, it was 19mm.<br />

Complications were evenly distributed between the<br />

two groups. The conclusion was that the advancement<br />

achieved with distraction is superior to that of<br />

standard Le Fort III. A halo distractor was preferred<br />

over the internal device because of better vector<br />

control. Focusing the vector on the facial midline<br />

allowed better correction of the concave facial profile<br />

to a convex profile.<br />

Cohen’s 485 series of 11 patients treated with the<br />

Modular <strong>Internal</strong> Distraction System (MIDS)<br />

(Howmedica-Leibinger) included 4 Le Fort III cases<br />

47


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

and 3 monobloc advancements. Midface retrusion<br />

and exorbitism were corrected in all patients and<br />

there were no infections in the monobloc group.<br />

Nadal 513 reported similar results in 8 patients who<br />

had monobloc advancement by distraction. Cohen 514<br />

also published a case report of a patient with facial<br />

bipartition treated with the MIDS.<br />

To avoid extensive surgery for removal of the<br />

device, Cohen and Holmes 487 used resorbable<br />

Macropore mesh as a substitute for the metallic fixation<br />

plate and achieved 20mm of distraction at the<br />

Le Fort III level in a 4-year-old girl with Crouzon<br />

syndrome.<br />

Distraction has also been described in Le Fort II<br />

advancement, 515 nasal bone lengthening, 516<br />

zygoma, 487,517 alveolar clefts, 518 and craniosynostosis.<br />

463,519,520<br />

Reviews and Long-Term Follow-Up<br />

Swennen and colleagues, 489 in an attempt to quantify<br />

clinical indications and treatment protocols,<br />

reviewed the literature on DO from 1966 to<br />

December 1999. They found 285 articles, including<br />

109 clinical studies involving treatment of 828<br />

patients. This impressive review provides an insight<br />

into the most widely employed indications and protocols.<br />

Mandibular distraction: Of 579 patients, 74.3%<br />

underwent mandibular lengthening. A latency period<br />

of 5–7 days was most common, as were a distraction<br />

rate of 1mm/day, with rhythms between 1 and 4<br />

times per day and a 6–8 week consolidation period.<br />

Unidirectional devices were used in 80.2% of cases,<br />

bidirectional in 15.6%, and multidirectional in 4.2%.<br />

External devices were used in 72% of cases and<br />

internal in 18%. Lengthening varied from 1–95mm.<br />

Maxillary distraction: Of 129 patients, 122<br />

underwent maxillary advancement. An external<br />

device was used in 95% of cases and an internal<br />

device in 5%. The average latency was 4–5 days, the<br />

rate was 1mm/day, and consolidation of 2–3 months<br />

was most common. Distraction lengths were 1–<br />

17mm.<br />

Simultaneous mandibular and maxillary distraction:<br />

This group included 24 patients who underwent<br />

mandibular DO with simultaneous maxillary<br />

48<br />

DO though IMF, most often though unilateral mandibular<br />

DO and an associated Le Fort I osteotomy.<br />

Mandibular distraction was accomplished with an<br />

external device in 87.5%, an internal device in the<br />

remaining 12.5%. Mandibular lengthening varied<br />

from 12–18mm.<br />

Midface and cranium: Of 96 cases, 64.6% had<br />

Le Fort III advancement and 35.4% had monobloc<br />

advancement. In patients younger than 4 years,<br />

monobloc was the most common procedure.<br />

The researchers’ conclusions were as follows:<br />

1. The issue of overcorrection remains controversial.<br />

2. A rate of 1mm/day is average.<br />

3. The standard latency period of 5–7 days could<br />

possibly be shortened, given the difference<br />

between the long bones and the membranous<br />

bones of the cranium and face.<br />

4. The complication rate was 22%, though most of<br />

these were mechanical problems associated with<br />

the device or minor pin site infections.<br />

5. There is a lack of long term data, especially<br />

regarding relapses.<br />

6. More objective data are necessary to refine protocols.<br />

7. The future of DO probably involves the development<br />

of miniaturized, multidirectional, and<br />

motorized devices.<br />

Mofid and colleagues 521 reviewed 3278 cases of<br />

craniofacial DO by means of a 4-page survey sent to<br />

2476 craniofacial and oral/maxillofacial surgeons<br />

worldwide. The response rate was 11.4%. Relapse<br />

was recognized by 50.4% of respondents; of these,<br />

64.8% had mandibular relapse and 60.6% had<br />

midface relapse. Relapse occurred within 6 months<br />

of completion of distraction in more than 66%. In<br />

cases of mandibular distraction, a higher use of internal<br />

devices (24.3%) and a higher rate (33.9%) of<br />

multivector devices was seen than reported by<br />

Swennen. 489 This most likely reflects advances in<br />

device design and surgeon experience. As noted by<br />

Swennen, the average latency was 4.9 days, the commonest<br />

rate was 1mm/day, and the average consolidation<br />

period was 7 weeks: 6 days in the mandible<br />

and 8 weeks and 6 days in the midface. Average


lengthening was 16.8mm in the mandible and<br />

14.5mm in the midface.<br />

Other complications of craniofacial distraction<br />

osteogenesis are tabulated in Table 8. 521<br />

AUGMENTATION AND CONTOURING PROCEDURES<br />

There are many indications for autogenous bone<br />

grafts in craniofacial surgery. 522 Most craniofacial<br />

surgeons believe that autogenous grafts of bone or<br />

cartilage are the materials of choice in craniofacial<br />

reconstruction because of their resistance to infection<br />

and low morbidity compared with alloplastic<br />

implants. 523 For an in-depth review of the subject,<br />

the reader is referred to Selected Readings in <strong>Plastic</strong><br />

<strong>Surgery</strong> volume 10, number 2. 524<br />

The calvarium is usually the preferred source of<br />

donor autogenous bone for grafting in the craniofacial<br />

skeleton. 79,80,525,526 Cutting 525 delineated the vascular<br />

supply to the cranium and introduced the concept<br />

of using vascularized calvarial bone in craniofa-<br />

TABLE 8<br />

Complication Rates<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

cial procedures. He determined that the most<br />

important source of perfusion to the cranium is the<br />

middle meningeal artery, which of course is not useful<br />

as a pedicle for bone transfer. Branches of the<br />

anterior and posterior deep temporal arteries, which<br />

also supply the temporalis muscle, constitute a lesser<br />

vascular supply to the cranium. Finally there is the<br />

vascular plexus fed by the supraorbital, supratrochlear,<br />

superficial temporal, and occipital arteries. The<br />

temporoparietalis fascia (superficial temporal fascia)<br />

contains the superficial temporal artery and provides<br />

a clinically useful pedicle to support a vascularized<br />

calvarial bone graft. 525,527<br />

McCarthy and colleagues 79,80 suggest leaving the<br />

galea and overlying vascular network broadly<br />

attached to the bone when transferring a vascularized<br />

calvarium bone flap because of the poor vascular<br />

connections within the periosteum.<br />

A review of vascularized calvarial inlay grafts based<br />

on a temporalis myoosseous design 528 notes preservation<br />

of the growth potential and a patent suture.<br />

(Reprinted with permission from Mofid MM, Manson PN, Robertson BC, et al: <strong>Craniofacial</strong> distraction osteogenesis: a review of 3278<br />

cases. Plast Reconstr Surg 108:1103, 2001.)<br />

49


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

Transsutural growth in the vascularized grafts continues<br />

for the duration of maxillary development despite cessation<br />

of skeletal growth at the calvarial donor site.<br />

Byrd and Hobar 529 described the use of porous<br />

hydroxyapatite (HA) granules in craniofacial augmentation<br />

procedures when structural support is<br />

not the main consideration. The HA granules contain<br />

pores approximately 200µ in diameter which<br />

allow fibrovascular ingrowth, so the implants are<br />

vascularized within a very few weeks. The HA<br />

granules are mixed with blood and microfibrillar<br />

collagen to make them easier to work with, and<br />

are then injected into a subperiosteal pocket wherever<br />

augmentation is desired. Onlays of hydroxyapatite<br />

do not resorb over time, are very seldom<br />

associated with infection, and seem to provoke no<br />

tissue reaction. The authors’ experience now<br />

extends to more than 200 patients operated on<br />

over an 8-year period, and they remain enthusiastic<br />

about the technique.<br />

A powdered form of hydroxyapatite marketed<br />

under the name of Bone Source (Leibinger Corp,<br />

Carrollton, Texas) has found application in craniofacial<br />

contour refinement. Burnstein and coworkers 530<br />

describe their experience with 61 patients, 56 of<br />

them children, treated with Bone Source for secondary<br />

craniofacial reconstruction over a 3- year period.<br />

The authors note excellent contour and volume<br />

retention and no interference with craniofacial<br />

growth.<br />

Jackson 531 remarks that the smaller pore size of<br />

the powdered HA compared with HA granules lends<br />

itself to a higher rate of bony replacement. They<br />

achieved excellent results in 20 patients, yet caution<br />

that long-term follow-up is necessary to establish the<br />

safety and reliability of its use.<br />

SURGERY FOR RADIATION-INDUCED DEFORMITY<br />

Jackson and colleagues 532 studied 14 children who<br />

received therapeutic radiation for malignant tumors<br />

in the orbital area and who subsequently showed<br />

widespread craniofacial deformities. The original<br />

tumors were retinoblastoma, rhabdomyosarcoma,<br />

and embryonic cell carcinoma. Years later, maldevelopment<br />

of the skull, orbit, maxilla, and mandible<br />

became apparent. The authors recommend correction<br />

of four levels of skeletal deformity in a single<br />

procedure, as follows:<br />

50<br />

1. frontotemporal expansion with repositioning of<br />

the cranial base<br />

2. orbital expansion and repositioning<br />

3. maxillary surgery with bone grafts to the zygoma<br />

as required<br />

4. mandibular lengthening and repositioning<br />

Bone grafts should be inlay rather than onlay, and<br />

soft tissue should be supplied by free-tissue transfer.<br />

At a second operation, the eye socket and eyelids<br />

are reconstructed to allow more satisfactory rehabilitation<br />

with an ocular prosthesis. On the basis of<br />

observations made during extensive skull base and<br />

orbital dissections, Jackson and colleagues 532 believe<br />

the radiation impaired growth of the sphenoid, which<br />

in turn locked the upper face and prevented normal<br />

development of the facial skeleton. In addition, the<br />

frontal, ethmoid, and maxillary sinuses failed to<br />

expand, resulting in further decrease of craniofacial<br />

dimensions. Craniotomy is essential to position the<br />

skull base correctly in order to accurately seat and<br />

remodel the orbit and maxilla. For further reading<br />

on this subject, one should refer to the articles by<br />

Nwoku, 533 Larson, 534 Marx, 535 Guyuron, 536,537 and<br />

Kawamoto. 538<br />

Cohen, Bartlett, and Whitaker 335 described their<br />

experience with reconstruction of late craniofacial<br />

deformities after therapeutic radiation of the head<br />

and face during childhood. High-dose radiation is a<br />

standard form of treatment for some pediatric craniofacial<br />

tumors, including retinoblastoma, rhabdomyosarcoma,<br />

Ewing’s sarcoma, and neurofibrosarcoma.<br />

The patients subsequently develop soft-tissue and<br />

bony hypoplasia of the irradiated areas, which can<br />

be partially corrected by onlay bone grafting and<br />

soft-tissue reconstruction with local pedicled flaps and<br />

dermal-fat grafts in multiple stages. 335 Forty percent<br />

of patients in their series required secondary procedures<br />

for improvement. The authors anticipate<br />

increasing usage of free-tissue transfer in these cases.<br />

Individuals who as children were irradiated for<br />

hemangiomas of the face are also presenting to plastic<br />

surgery clinics with severe craniofacial deformities.<br />

At one time high-beam radiotherapy was advocated<br />

for capillary hemangioma of the cheek, and<br />

the sequelae of this treatment in the growing child is<br />

now painfully evident as terrible facial deformities in<br />

the adult. Reconstruction should follow the principles<br />

outlined by Jackson and colleagues 532 above.


1. Sperber GH: <strong>Craniofacial</strong> Embryology, 4 th Ed. London,<br />

Wright, 1989.<br />

2. Johnston MC: Embryology of the Head and Neck. In:<br />

McCarthy JG (ed), <strong>Plastic</strong> <strong>Surgery</strong>. Philadelphia, WB<br />

Saunders, 1990. Vol 4, Ch 46, pp 2451-2495.<br />

3. Slavkin HC et al: Early embryonic mouse mandibular<br />

morphogenesis and cytodifferentiation in serumless, chemically<br />

defined medium: a model for studies of autocrine<br />

and/or paracrine regulatory factors. J Craniofac Genet Dev<br />

Biol 9:185, 1989.<br />

4. Slavkin HC: Developmental <strong>Craniofacial</strong> Biology. Philadelphia,<br />

Lea & Febiger, 1979.<br />

5. Cohen MM Jr, Rollnick BR, and Kaye CI: Oculoauriculovertebral<br />

spectrum: An updated critique. Cleft<br />

Palate J 26:276, 1989.<br />

6. Prescott NJ, Malcolm S: Folate and the face: evaluating<br />

the evidence for the influence of folate genes on craniofacial<br />

development. Cleft Palate Craniofac J 39:327,<br />

2002.<br />

7. Kallen K: Maternal smoking and craniosynostosis. Teratology<br />

60:146, 1999.<br />

8. Honein MA, Rasmussen SA, Reefhuis J, et al: Maternal<br />

smoking and environmental tobacco smoke exposure<br />

and the risk of orofacial clefts. Epidemiology 18:226,<br />

2007.<br />

9. Gardner JS, Guyard-Boileau B, Alderman BW, et al: Maternal<br />

exposure to prescription and non-prescription pharmaceuticals<br />

or drugs of abuse and risk of craniosynostosis.<br />

Int J Epidemiol 27:64, 1998.<br />

10. Whitaker LA, Pashayan H, Reichman J: A proposed new<br />

classification of craniofacial anomalies. Cleft Palate J<br />

18:161, 1981.<br />

11. Tessier P: Anatomical classification of facial, craniofacial<br />

and latero-facial clefts. J Maxillofac Surg 4:69, 1976.<br />

12. David DJ, Moore MH, Cooter RD: Tessier clefts revisited<br />

with a third dimension. Cleft Palate J 26:163, 1989.<br />

13. Van der Meulen JC et al: A morphogenetic classification of<br />

craniofacial malformations. Plast Reconstr Surg 71:560,<br />

1983.<br />

14. DeMyer W, Zeman W, Palmer CG: The face predicts the<br />

brain: Diagnostic significance of median facial anomalies<br />

for holoprosencephaly (arhinencephaly). Pediatrics 34:256,<br />

1964.<br />

15. Cohen MM Jr, Sulik KK: Perspectives on holoprosencephaly:<br />

<strong>Part</strong> II. Central nervous system, craniofacial anatomy,<br />

syndrome commentary, diagnostic approach, and experimental<br />

studies. J Craniofac Gen Dev Biol 12(4):196, 1992.<br />

16. Elias DL, Kawamoto HK Jr, Wilson LF: Holoprosencephaly<br />

and midline facial anomalies: Redefining classification and<br />

management. Plast Reconstr Surg 90:951, 1992.<br />

17. Dursy E: Zur Entwicklungsgeschichte des Kopfes des<br />

Menschen und der hoeheren Wirbelthiere. Tubingen,<br />

Verlag der H Lauppschen- Buchhandlung, 1869, p 99.<br />

18. His W: Die Entwickelung der menschlichen und t.ierischer<br />

Physiognomen. Arch Anat Entwicklungsgesch, 1892, p<br />

384.<br />

19. Kawamoto HK: The kaleidoscopic world of rare craniofacial<br />

clefts: order out of chaos (Tessier classification). Clin<br />

Plast Surg 3:529, 1976.<br />

BIBLIOGRAPHY<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

20. Pohlmann EH: Die embryonale Metamorphose der<br />

Physiognomie und der Mundhoehle des Katzenkopfes.<br />

Morphol Jahrb Leipzig 41:617, 1910.<br />

21. Veau V: Hasencharten menschlicher Keimlinge auf der<br />

Stufe 21-23 mm SSL. Z Anat Entwicklungsgesch 108:459,<br />

1938.<br />

22. Stark RB: The pathogenesis of harelip and cleft palate. Plast<br />

Reconstr Surg 13:20, 1954.<br />

23. Stark RB, Saunders DE: The first branchial syndrome: The<br />

oral-mandibular-auricular syndrome. Plast Reconstr Surg<br />

29:299, 1962.<br />

24. Pantaloni M and Byrd HS, Salomon J, and Hollier L: Cleft<br />

Lip I: Primary Deformities; Cleft Lip II: Secondary Deformities;<br />

and Cleft Palate and Velopharyngeal Incompetence.<br />

Selected Read Plast Surg 9(21-23), 2001.<br />

25. Ozaki W, Kawamoto HK Jr: <strong>Craniofacial</strong> Clefting. In: Lin<br />

KY, Ogle RC, Jane JA (eds), <strong>Craniofacial</strong> <strong>Surgery</strong>. Science<br />

and Surgical Technique. Philadelphia, Saunders, 2002,<br />

Ch 21.<br />

26. Van der Meulen JCH: Oblique facial clefts: Pathology,<br />

etiology, and reconstruction. Plast Reconstr Surg 76:212,<br />

1985.<br />

27. Resnick JI, Kawamoto HK Jr: Rare craniofacial clefts:<br />

Tessier No. 4 clefts. Plast Reconstr Surg 85:843, 1990.<br />

28. Galante G, Dado DV: The Tessier number 5 cleft: A report<br />

of two cases and a review of the literature. Plast Reconstr<br />

Surg 88:131, 1991.<br />

29. Fuente del Campo A: Surgical correction of Tessier number<br />

8 cleft. Plast Reconstr Surg 86:658, 1990.<br />

30. Menard RM, Moore MH, David DJ: Tissue expansion in<br />

the reconstruction of Tessier craniofacial clefts: a series of<br />

17 patients. Plast Reconstr Surg 103:779, 1999.<br />

31. Thomson A: As cited in Grabb WC: The first and second<br />

branchial arch syndrome. Plast Reconstr Surg 36:485,<br />

1965.<br />

32. Posnick JC: Hemifacial microsomia: evaluation and<br />

staging of reconstruction. J Oral Maxillofac Surg 56:642,<br />

1998.<br />

33. Posnick JC: <strong>Craniofacial</strong> and Maxillofacial <strong>Surgery</strong> in<br />

Children and Young Adults. Philadelphia, WB Saunders,<br />

2000.<br />

34. Bassila M, Goldberg R: The association of facial palsy and<br />

sensorineural hearing loss in patients with hemifacial microsomia.<br />

Cleft Palate J 26:287, 1989.<br />

35. Poswillo DE: The pathogenesis of the first and second<br />

branchial arch syndrome. Oral Surg 35:302, 1973.<br />

36. Converse JM et al: Bilateral facial microsomia: Diagnosis,<br />

classification, and treatment. Plast Reconstr Surg 59:653,<br />

1977.<br />

37. McKenzie J, Craig J: Mandibulofacial dysostosis. Arch Dis<br />

Child 30:391, 1952.<br />

38. Johnston MC, Sulik KK: Embryology of the head and neck.<br />

In: Serafin D and Georgiade NG (eds), Pediatric <strong>Plastic</strong><br />

<strong>Surgery</strong>. St Louis, CV Mosby, 1984. Vol 1, Ch 13, pp 184-<br />

215.<br />

39. Murray JE, Kaban LB, Mulliken JB, Evans CA: Analysis and<br />

treatment of hemifacial microsomia. In: Caronni EP (ed),<br />

<strong>Craniofacial</strong> <strong>Surgery</strong>. Boston, Little Brown, 1982. Ch 33,<br />

pp 377- 390.<br />

51


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

40. Polley JW, Figueroa AA, LIOU EJW, Cohen M: Longitudinal<br />

analysis of mandibular asymmetry in hemifacial microsomia.<br />

Plast Reconstr Surg 99:328, 1997.<br />

41. Kearns GJ, Padwa BL, Mulliken JB, Kaban LB: Progression<br />

of facial asymmetry in hemifacial microsomia. Plast Reconstr<br />

Surg 105:492, 2000.<br />

42. Munro IR: Treatment of craniofacial microsomia. Clin<br />

Plast Surg 14:177, 1987.<br />

43. Murray JE, Kaban LB, Mulliken JB: Analysis and treatment<br />

of hemifacial microsomia. Plast Reconstr Surg 74:186,<br />

1984.<br />

44. Munro IR: One-stage reconstruction of the temporomandibular<br />

joint in hemifacial microsomia. Plast Reconstr Surg<br />

66:699, 1980.<br />

45. Pruzansky S: Not all dwarfed mandibles are alike. Birth<br />

Defects 5:120, 1969.<br />

46. Meurman Y: Congenital microtia and meatal atresia. Arch<br />

Otolaryngol 66:443, 1957.<br />

47. Mulliken JB, Kaban LB: Analysis and treatment of hemifacial<br />

microsomia in childhood. Clin Plast Surg 14:91, 1987.<br />

48. Bennun RD et al: Microtia: A microform of hemifacial<br />

microsomia. Plast Reconstr Surg 76:859, 1985.<br />

49. David DJ, Mahatumarat C, Cooter RD: Hemifacial microsomia:<br />

a multisystem classification. Plast Reconstr Surg<br />

80:525, 1987.<br />

50. Posnick JC: Surgical correction of mandibular hypoplasia<br />

in hemifacial microsomia: a personal perspective. J Oral<br />

Maxillofac Surg 56:639, 1998.<br />

51. Converse JM et al: The corrective treatment of the skeletal<br />

asymmetry in hemifacial microsomia. Plast Reconstr Surg<br />

52:221, 1973.<br />

52. Ortiz-Monasterio F: Early mandibular and maxillary osteotomies<br />

for the correction of hemifacial microsomia: A<br />

preliminary report. Clin Plast Surg 9:509, 1982.<br />

53. Munro IR, Lauritzen CGK: Classification and treatment of<br />

hemifacial microsomia. In: Caronni EP (ed), <strong>Craniofacial</strong><br />

<strong>Surgery</strong>. Boston, Little Brown, 1984. Ch 34, pp 391-400.<br />

54. Ortiz-Monasterio F, Fuente del Campo A: Early skeletal<br />

correction of hemifacial microsomia. In: Caronni EP (ed),<br />

<strong>Craniofacial</strong> <strong>Surgery</strong>. Boston, Little Brown, 1984. Ch 35,<br />

pp 401- 410.<br />

55. Moss ML: The functional matrix. In: Kraus BS, Reidel RA<br />

(eds), Vistas in Orthodontics. Philadelphia, Lea & Febiger,<br />

1962.<br />

56. Moss ML et al: The passive role of nasal septal cartilage in<br />

mid-facial growth. Plast Reconstr Surg 41:536, 1968.<br />

57. McCarthy JG et al: Lengthening the human mandible by<br />

gradual distraction. Plast Reconstr Surg 89:1, 1992.<br />

58. Molina F, Ortiz Monasterio F: Mandibular elongation and<br />

remodeling by distraction: a farewell for major osteotomies.<br />

Plast Reconstr Surg 96:825, 1995.<br />

59. McCarthy JG et al: <strong>Craniofacial</strong> Microsomia. In: McCarthy<br />

JG (ed), <strong>Plastic</strong> <strong>Surgery</strong>. Philadelphia, WB Saunders,<br />

1990. Vol 4, Ch 62, pp 3054-3100.<br />

60. Ross RB: Costochondral grafts replacing the mandibular<br />

condyle. Cleft Palate Craniofac J 36:334, 1999.<br />

61. La Rossa D, Whitaker L, Dabb R, Mellissinos E: The use of<br />

microvascular free flaps for soft tissue augmentation of the<br />

face in children with hemifacial microsomia. Cleft Palate<br />

J 17:138, 1980.<br />

62. Upton J et al: Restoration of facial contour using free<br />

vascularized omental transfer. Plast Reconstr Surg 66:560,<br />

1980.<br />

52<br />

63. Janis JE, Leedy JE: Lip, Cheek and Scalp Reconstruction.<br />

Selected Read Plast Surg 10(13), 2006.<br />

64. Rogers BO: Berry-Treacher Collins syndrome: A review<br />

of 200 cases. Br J Plast Surg 17:109, 1964.<br />

65. Munro IR et al: <strong>Craniofacial</strong> Syndromes. In: McCarthy JG<br />

(ed), <strong>Plastic</strong> <strong>Surgery</strong>. Philadelphia, WB Saunders, 1990.<br />

Vol 4, Ch 63, pp 3101-3123.<br />

66. McCarthy JG, Zide BM: Rare craniofacial clefts. In: Serafin<br />

D and Georgiade NG (eds), Pediatric <strong>Plastic</strong> <strong>Surgery</strong>. St<br />

Louis, CV Mosby, 1984. Vol 1, Ch 25, p 390.<br />

67. Tessier P: Surgical correction of Treacher Collins syndrome.<br />

In: Bell WH (ed), Modern Practice in Orthognathic<br />

and Reconstructive <strong>Surgery</strong>. Philadelphia, Saunders,<br />

1992. Vol 2, p 1601.<br />

68. Dixon M, Read A, Donnai D, et al: The gene for Treacher<br />

Collins syndrome maps to the long arm of chromosome 5.<br />

Am J Hum Genet 49:17, 1991.<br />

69. Jabs E, Li X, Coss C, et al: Mapping the Treacher Collins<br />

syndrome locus to 5q31.3®q33.3 Genomics 11:193,<br />

1991.<br />

70. Marsh KL, Dixon J, Dixon MJ: Mutations in the Treacher<br />

Collins syndrome gene lead to mislocalization of the nucleolar<br />

protein treacle. Hum Mol Genet 7:1795, 1998.<br />

71. Marszalek B, Wojcicki P, Kobus K, Trzeciak WH: Clinical<br />

features, treatment and genetic background of Treacher<br />

Collins syndrome. J Appl Genet 43:223, 2002.<br />

72. Sulik KK, JOhnston MC, Smiley SJ, et al: Mandibulofacial<br />

dysostosis (Treacher Collins syndrome): a new proposal for<br />

its pathogenesis. Am J Med Genet 27:245, 1987.<br />

73. Shprintzen RJ et al: Pharyngeal hypoplasia in Treacher<br />

Collins syndrome. Arch Otolaryngol 105:127, 1979.<br />

74. Behrents RG, McNamara JA, Avery JK: Prenatal<br />

mandibulofacial dysostosis (Treacher Collins syndrome).<br />

Cleft Palate J 14:13, 1977.<br />

75. Jackson IT: Reconstruction of the lower eyelid defect in<br />

Treacher Collins syndrome. Plast Reconstr Surg 67:365,<br />

1981.<br />

76. Raulo Y: Treacher Collins syndrome: Analysis and principles<br />

of surgery. In: Caronni EP (ed), <strong>Craniofacial</strong><br />

<strong>Surgery</strong>. Boston, Little Brown, 1984. Ch 32, pp 371-376.<br />

77. Posnick JC: Treacher Collins Syndrome. In: Aston S (ed),<br />

Grabb and Smith’s <strong>Plastic</strong> <strong>Surgery</strong>, 5th ed. Philadelphia,<br />

Lippincott Raven, 1997.<br />

78. Posnick JC, Goldstein JA, Waitzman AA: Surgical correction<br />

of the Treacher Collins malar deficiency: Quantitative<br />

CT scan analysis of long-term results. Plast Reconstr Surg<br />

92:12, 1993.<br />

79. McCarthy JG, Zide BM: The spectrum of calvarial bone<br />

grafting: Introduction of the vascularized calvarial bone<br />

flap. Plast Reconstr Surg 74:10, 1984.<br />

80. McCarthy JG, Cutting CB, Shaw WW: Vascularized calvarial<br />

flaps. Clin Plast Surg 14:37, 1987.<br />

81. Freihofer HPM: Variations in the correction of Treacher<br />

Collins syndrome. Plast Reconstr Surg 99:647, 1997.<br />

82. Van der Meulen JCH et al: The use of a temporal<br />

osteoperiosteal flap for the reconstruction of malar hypoplasia<br />

in Treacher Collins syndrome. Plast Reconstr Surg<br />

74:687, 1984.<br />

83. Psillakis JM et al: Vascularized outer-table calvarial bone<br />

flaps. Plast Reconstr Surg 78:309, 1986.<br />

84. Brent B: Auricular repair with autogenous rib cartilage<br />

grafts: two decades of experience with 600 cases. Plast<br />

Reconstr Surg 90:355, 1992.


85. Dufresne C: Treacher Collins Syndrome. In: Dufresne C<br />

(ed), Complex <strong>Craniofacial</strong> Problems: A Guide to<br />

Analysis and Treatment. New York, Churchill Livingstone,<br />

1992; p 281.<br />

86. Munro IR et al: <strong>Craniofacial</strong> Syndromes. In: McCarthy JG<br />

(ed), <strong>Plastic</strong> <strong>Surgery</strong>. Philadelphia, WB Saunders, 1990.<br />

Vol 4, Ch 63, pp 3101-3160.<br />

87. Jackson I et al: A significant feature of Nager’s syndrome:<br />

Palatal agenesis. Plast Reconstr Surg 84:219, 1989.<br />

88. Zuckerkandl E: Fossae praenasales. Normale und<br />

pathologische. Anat Nasenhohle 1(2):48, 1882.<br />

89. Noyes FB: Case report. Angle Orthod 9:160, 1939.<br />

90. Von Binder KH: Dysostosis maxillo-nasalis, ein<br />

arhinencephaler Missbildungskomplex. Deutsche<br />

Zahnaerztl Z 17:438, 1962.<br />

91. Munro IR, Sinclair WJ, Rudd NL: Maxillonasal dysplasia<br />

(Binder’s syndrome). Plast Reconstr Surg 63:657, 1979.<br />

92. Posnick JC, Tompson B: Binder syndrome: staging of<br />

reconstruction and skeletal stability and relapse patterns<br />

after Le Fort I osteotomy using miniplate fixation. Plast<br />

Reconstr Surg 99:961, 1997.<br />

93. Holmstrom H: Clinical and pathologic features of<br />

maxillonasal dysplasia (Binder’s syndrome): significance of<br />

the prenasal fossa on etiology. Plast Reconstr Surg 78:559,<br />

1986.<br />

94. Rival JM, Gherga-Negrea A, Mainard R, Delaire J: Dysostose<br />

maxillo-nasale de Binder. J Genet Hum 22:263, 1974.<br />

95. Olow-Nordenram M, Valentin J: An etiologic study of<br />

maxillonasal dysplasia: Binder’s syndrome. Scand J Dent<br />

Res 96:69, 1988.<br />

96. Holmstrom H: Surgical correction of the nose and midface<br />

in maxillonasal dysplasia (Binder’s syndrome). Plast Reconstr<br />

Surg 78:568, 1986.<br />

97. Jackson IT, Moos KF, Sharpe DT: Total surgical management<br />

of Binder’s syndrome. Ann Plast Surg 7:25, 1981.<br />

98. Banks P, Tanner B: The mask rhinoplasty: a technique for<br />

the treatment of Binder’s syndrome and related disorders.<br />

Plast Reconstr Surg 92:1038, 1993.<br />

99. Converse JM: Restoration of facial contour by bone grafts<br />

introduced through the oral cavity. Plast Reconstr Surg<br />

6:295, 1950.<br />

100. Converse JM, Horowitz SL, Valauri AJ, et al: The treatment<br />

of nasomaxillary hypoplasia: a new pyramidal nasoorbital<br />

maxillary osteotomy. Plast Reconstr Surg 45:527,<br />

1970.<br />

101. Ragnell A: Nasomaxillary retroposition in children. Successive<br />

reconstruction of the nose: a preliminary report.<br />

Nord Med 77:847, 1967.<br />

102. Henderson D, Jackson IT: Naso-maxillary hypoplasia: the<br />

Le Fort II osteotomy. Br J Oral Surg 11:77, 1973.<br />

103. Steinhauser EW: Variations of Le Fort II osteotomies for<br />

correction of midfacial deformities. J Maxillofac Surg<br />

8:258, 1980.<br />

104. Tessier P, Tulasne JF, Delaire J, et al: Therapeutic aspects<br />

of maxillonasal dysostosis (Binder syndrome). Head Neck<br />

Surg 3:207, 1981.<br />

105. Holmstrom H, Kahnberg KE: Surgical approach in severe<br />

cases of maxillonasal dysplasia (Binder’s syndrome). Swed<br />

Dent J 12:3, 1988.<br />

106. Rune B, Aberg M: Bone grafts to the nose in Binder’s<br />

syndrome (maxillonasal dysplasia): a follow-up of eleven<br />

patients with the use of profile roentgenograms. Plast<br />

Reconstr Surg 101:297, 1998.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

107. Wolfe SA: Lengthening the nose: a lesson from craniofacial<br />

surgery applied to posttraumatic and congenital deformities.<br />

Plast Reconstr Surg 94:78, 1994.<br />

108. McCollum AGH, Wolford LM: Binder syndrome: literature<br />

review and long-term follow-up on two cases. Int J<br />

Adult Orthod Orthognath Surg 13:45, 1998.<br />

109. Robin P: Backward fall of the root of the tongue as cause<br />

of respiratory disturbances. Bull Acad Med (Paris) 89:37,<br />

1923.<br />

110. Fletcher MM, Blum SL, Blanchard CL: Pierre Robin<br />

syndrome: pathophysiology of obstructive episodes. Laryngoscope<br />

79:547, 1969.<br />

111. Kiskadden WS, Dietrich SR: Review of the treatment of<br />

micrognathia. Plast Reconstr Surg 12:365, 1953.<br />

112. Douglas B: The treatment of macrognathia associated with<br />

obstruction by a plastic procedure. Plast Reconstr Surg<br />

1:300, 1946.<br />

113. Randall P: The Robin sequence: Micrognathia and<br />

glossoptosis with airway obstruction. In: McCarthy JG (ed),<br />

<strong>Plastic</strong> <strong>Surgery</strong>. Philadelphia, WB Saunders, 1990. Vol 4,<br />

Ch 63, pp 3123-3134.<br />

114. Pashayan HM, Lewis MB: Clinical experience with the<br />

Robin sequence. Cleft Palate J 21:270, 1984.<br />

115. Routledge RT: The Pierre Robin syndrome: A surgical<br />

emergency in the neonatal period. Br J Plast Surg 13:204,<br />

1960.<br />

116. Denny AD, Talisman R, Hanson PR, Recinos RF: Mandibular<br />

distraction osteogenesis in very young patients to correct<br />

airway obstruction. Plast Reconstr Surg 108:302,<br />

2001.<br />

117. Jackson IT, Tanner NSB, Hide TAH: Frontonasal encephalocele—“long<br />

nose hypertelorism”. Ann Plast Surg 11:490,<br />

1983.<br />

118. Simpson DA, David DJ, White J: Cephaloceles: treatment,<br />

outcome, and antenatal diagnosis. Neurosurgery 15:14,<br />

1984.<br />

119. Suwanwela C, Suwanwela N: A morphological classification<br />

of scincipital encephalomeningoceles. J Neurosurg<br />

36:201, 1972.<br />

120. Mazzola RF: Congenital malformations in the frontonasal<br />

area: Their pathogenesis and classification. Clin Plast Surg<br />

3(4):573, 1976.<br />

121. David DJ et al: Fronto-ethmoidal meningoencephaloceles:<br />

morphology and treatment. Br J Plast Surg 37:271, 1984.<br />

122. Jacob OJ, Rosenfeld JV, Watters DAK. The repair of frontal<br />

encephaloceles in Papua New Guinea. Aust N Z J Surg<br />

64:856, 1994.<br />

123. Cohen MM: Craniosynostosis and syndromes with craniosynostosis:<br />

incidence, genetics, penetrance, variability,<br />

and new syndrome updating. Birth Defects 15 (5B):13,<br />

1979.<br />

124. Humphreys RP: Encephalocele and dermal sinuses. In:<br />

Cheek WR, Marlin AE, McLone DG, et al (eds), Pediatric<br />

Neurosurgery: <strong>Surgery</strong> of the Developing Nervous<br />

System, 3rd ed. Philadelphia, WB Saunders, 1994, p 96.<br />

125. Charoonsmith T, Suwanwela C: Frontoethmoidal<br />

encephalomeningocele with special reference to plastic<br />

reconstruction. Clin Plast Surg 1:27, 1974.<br />

126. Pollack IF: Management of encephaloceles and craniofacial<br />

problems in the neonatal period. Neurosurg Clin<br />

North Am 9:121, 1998.<br />

127. David DJ: Cephaloceles: Classification, pathology, and<br />

management. A review. J Craniofac Surg 4:192, 1993.<br />

53


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

128. Forcada M, Montandon D, Rilliet B: Frontoethmoidal<br />

cephaloceles: Transcranial and transfacial surgical treatment.<br />

J Craniofac Surg 4:203, 1993.<br />

129. Smit CSF et al: Frontoethmoidal meningoencephaloceles:<br />

A review of 14 consecutive patients. J Craniofac Surg<br />

4:210, 1993.<br />

130. Holmes AD, Meara JG, Kolker AR, et al: Frontoethmoidal<br />

encephaloceles: reconstruction and refinements. J<br />

Craniofac Surg 12:6, 2001.<br />

131. Persing JA, Jane JA, Shaffrey M: Virchow and the pathogenesis<br />

of craniosynostosis: A translation of his original work.<br />

Plast Reconstr Surg 83:738, 1989.<br />

132. Alden TD, Lin KY, Jane JA: Mechanisms of premature<br />

closure of cranial sutures. Child Nerv Syst 15:670, 1999.<br />

133. Robin NH: Molecular genetic advances in understanding<br />

craniosynostosis. Plast Reconstr Surg 103:1060, 1999.<br />

134. Shillito J, Matson DD: Craniosynostosis: A review of 519<br />

surgical patients. Pediatrics 41:829, 1968.<br />

135. Shuper A et al: The incidence of isolated craniosynostosis<br />

in the newborn infant. Am J Dis Child 139:85, 1985.<br />

136. Marchac D, Renier D: <strong>Craniofacial</strong> <strong>Surgery</strong> for Craniosynostosis.<br />

Boston, Little Brown, 1982.<br />

137. Oakes WJ: Craniosynostosis. In: Serafin D, Georgiade NG<br />

(eds), Pediatric <strong>Plastic</strong> <strong>Surgery</strong>. St Louis, CV Mosby,<br />

1984. Vol 1, Ch 26, pp 404-439.<br />

138. Cohen MM Jr, MacLean RE: Anatomic, Genetic, Nosologic,<br />

Diagnostic, and Psychosocial Considerations. In:<br />

Cohen MM Jr, MacLean RE (eds), Craniosynostosis. Diagnosis,<br />

Evaluation, and Management, 2nd Ed. New York,<br />

Oxford Univ Press, 2000, Ch 11, p 119.<br />

139. Huang MHS, Gruss JS, Clarren SK, et al: The differential<br />

diagnosis of posterior plagiocephaly: true lambdoid synostosis<br />

versus positional molding. Plast Reconstr Surg 98:765,<br />

1996.<br />

140. American Academy of Pediatrics Task Force on Infant<br />

Positioning and SIDS: Positioning and SIDS. Pediatrics<br />

89:1120, 1989.<br />

141. Losee JE, Mason AC: Deformational plagiocephaly: diagnosis,<br />

prevention, and treatment. Clin Plast Surg 32:53,<br />

2005.<br />

142. Vies JS, Colla C, Weber JW, et al: Helmet versus nonhelmet<br />

treatment in non-synostotic positional posterior plagiocephaly.<br />

J Craniofac Surg 11:572, 2000.<br />

143. Latham RA: Maxillary development and growth: The<br />

septopremaxillary ligament. J Anat 107:471, 1970.<br />

144. Latham RA: Growth and development of the craniofacial<br />

skeleton. In: Serafin D and Georgiade NG (eds), Pediatric<br />

<strong>Plastic</strong> <strong>Surgery</strong>. St Louis, CV Mosby, 1984. Vol 1, Ch 14,<br />

pp 216-229.<br />

145. Moss ML: The pathogenesis of premature cranial synostosis<br />

in man. Acta Anat (Basel) 37:351, 1959.<br />

146. Cohen MM Jr: Sutural Pathology. In: Cohen MM Jr,<br />

MacLean RE (eds), Craniosynostosis. Diagnosis, Evaluation,<br />

and Management, 2nd Ed. New York, Oxford Univ<br />

Press, 2000, Ch 5.<br />

147. Albright AL, Byrd RP: Suture pathology in craniosynostosis.<br />

J Neurosurg 54:384, 1981.<br />

148. Graham JM Jr, deSaxe M, Smith DW: Sagittal craniostenosis:<br />

fetal head constraint as one possible cause. J Pediatr<br />

95(5 Pt 1):747, 1979.<br />

149. Graham JM Jr, Badura RJ, Smith DW: Coronal craniostenosis:<br />

fetal head constraint as one possible cause. Pediatrics<br />

65:995, 1980.<br />

54<br />

150. Kloss JL: Craniosynostosis secondary to ventriculoatrial<br />

shunt. Am J Dis Child 116:315, 1968.<br />

151. Shunts and problems in shunts. Monogr Neural Sci 8:1-<br />

228, 1982.<br />

152. Noetzel MJ et al: Hydrocephalus and mental retardation in<br />

craniosynostosis. J Pediatr 107:885, 1985.<br />

153. Robinson DC, Hall R, Munro DS: Graves disease. An unusual<br />

complication: raised intracranial pressure due to premature<br />

fusion of skull sutures. Arch Dis Child 44:252, 1969.<br />

154. Breugem CC, van R Zeeman BJ: Retrospective study of<br />

nonsyndromic craniosynostosis treated over a 10-year<br />

period. J Craniofac Surg 10:140, 1999.<br />

155. Cohen SR, Dauser RC, Gorski JL: Insidious onset of familial<br />

craniosynostosis. Cleft Palate Craniofac J 30:401, 1993.<br />

156. Opperman LA, Sweeney TM, Redmon J, et al: Tissue<br />

interactions with underlying dura mater inhibit osseous<br />

obliteration of developing cranial sutures. Dev Dyn 198:312,<br />

1993.<br />

157. Opperman LA, Passarelli RW, Morgan EP, et al: Cranial<br />

sutures require tissue interactions with dura mater to resist<br />

osseous obliteration in vitro. J Bone Miner Res 10:1978,<br />

1995.<br />

158. Hobar PC et al: The role of the dura in cranial bone<br />

regeneration in the immature animal. Plast Reconstr Surg<br />

92:405, 1993.<br />

159. Hobar PC, Masson JA, Wilson R, Zerwekh J: The importance<br />

of the dura in craniofacial surgery. Plast Reconstr<br />

Surg 98:217, 1996.<br />

160. Greenwald JA, Mehrara BJ, Spector JA, et al: Biomolecular<br />

mechanisms of calvarial bone induction: immature versus<br />

mature dura mater. Plast Reconstr Surg 105:1382, 2000.<br />

161. Greenwald JA, Mehrara BJ, Spector JA, et al: Immature<br />

versus mature dura mater. II: Differential expression of<br />

genes critical to calvarial reossification. Plast Reconstr Surg<br />

106:630, 2000.<br />

162. Levine JP, Bradley JP, Roth DA, et al: Studies in cranial<br />

suture biology: regional dura mater determines overlying<br />

suture biology. Plast Reconstr Surg 101:1441, 1998.<br />

163. Opperman LA, Persing JA, Sheen R, Ogle RC: In the<br />

absence of periosteum, transplanted fetal and neonatal rat<br />

coronal sutures resist osseous obliteration. J Craniofac Surg<br />

5:327, 1994.<br />

164. Delezoide AL, Benoist-Lasselin C, Legeai-Mallet L, et al:<br />

Spatio-temporal expression of FGFR 1, 2 and 3 genes<br />

during human embryo-fetal ossification. Mech Dev 77:19,<br />

1998.<br />

165. Mangasarian K, Li Y, Mansukhani A, Basilico C: Mutation<br />

associated with Crouzon syndrome causes ligand-independent<br />

dimerization and activation of FGF receptor-2. J<br />

Cell Physiol 172:117, 1997.<br />

166. Most D, Levine JP, Chang J, et al: Studies in cranial suture<br />

biology: up-regulation of transforming growth factor-beta 1<br />

and basic fibroblast growth factor mRNA correlates with<br />

posterior frontal cranial suture fusion in the rat. Plast<br />

Reconstr Surg 101:1431, 1998.<br />

167. Mehrara BJ, Mackool RJ, McCarthy JG, et al: Immunolocalization<br />

of basic fibroblast growth factor and fibroblast<br />

growth factor receptor-1 and receptor-2 in rat cranial<br />

sutures. Plast Reconstr Surg 102:1805, 1998.<br />

168. Opperman LA, Nolen AA, Ogle RC: TGF-beta 1, TGF-beta<br />

2 and TGF-beta 3 exhibit distinct patterns of expression<br />

during cranial suture formation and obliteration in vivo and<br />

in vitro. J Bone Miner Res 12:301, 1997.


169. Opperman LA, Chhabra A, Cho RW, Ogle RC: Cranial<br />

suture obliteration is induced by removal of transforming<br />

growth factor (TGF)-beta 3 activity and prevented by<br />

removal of TGF-beta 2 activity from fetal rat calvaria in<br />

vitro. J Craniofac Genet Dev Biol 19:164, 1999.<br />

170. Mehrara BJ, Most D, Chang J, et al: Basic fibroblast growth<br />

factor and transforming growth factor beta-1 expression in<br />

the developing dura mater correlates with calvarial bone<br />

formation. Plast Reconstr Surg 104:435, 1999.<br />

171. Lin KY, Nolen AA, Gampper TJ, et al: Elevated levels of<br />

transforming growth factors beta 2 and beta 3 in lambdoid<br />

sutures from children with persistent plagiocephaly. Cleft<br />

Palate Craniofac J 34:331, 1997.<br />

172. Morriss-Kay GM, Iseki S, Johnson D: Genetic control of the<br />

cell proliferation-differentiation balance in the developing<br />

skull vault: roles of fibroblast growth factor receptor signalling<br />

pathways. Novartis Found Symp 232:102, 2001.<br />

173. Britto JA, Chan JCT, Evans RD, et al: Differential expression<br />

of fibroblast growth factor receptors in human digital development<br />

suggests common pathogenesis in complex<br />

acrosyndactyly and craniosynostosis. Plast Reconstr Surg<br />

107:1331, 2001.<br />

174. Cohen MM Jr: Discussion of “Differential expression of<br />

fibroblast growth factor receptors in human digital development<br />

suggests common pathogenesis in complex<br />

acrosyndactyly and craniosynostosis”, by Britto JA, Chan<br />

JCT, Evans RD, et al. Plast Reconstr Surg 107:1339, 2001.<br />

175. Jabs EW, Muller U, Li X, et al: A mutation in the<br />

homeodomain of the human MSX2 gene in a family affected<br />

with autosomal dominant craniosynostosis. Cell<br />

75:443, 1993.<br />

176. Vainio S, Karavanova I, Jowett A, Thesleff I: Identification<br />

of BMP-4 as a signal mediating secondary induction between<br />

epithelial and mesenchymal tissues during early<br />

tooth development. Cell 75:45, 1993.<br />

177. Graham A, Francis-West P, Brickell P, Lumsden A: The<br />

signalling molecule BMP4 mediates apoptosis in the<br />

rhomboencephalic neural crest. Nature 372:684, 1994.<br />

178. Amaya E, Musci TJ, Kirschner MW: Expression of a<br />

dominant negative mutant of the FGF receptor disrupts<br />

mesoderm formation in Xenopus embryos. Cell 66:257,<br />

1991.<br />

179. Preston A, Post J, Keats B, et al: A gene for Crouzon<br />

craniofacial dysostosis maps to the long arm of chromosome<br />

10. Nature Genet 7:149, 1994.<br />

180. Reardon W, Winter R, Rutland P, et al: Mutations in the<br />

fibroblast growth factor receptor 2 gene cause Crouzon<br />

syndrome. Nature Genet 8:98, 1994.<br />

181. Muenke M, Shell U, Hehr A, et al: A common mutation in<br />

the fibroblast growth factor receptor 1 gene in Pfeiffer<br />

syndrome. Nature Genet 8:269, 1994.<br />

182. Rutland P, Pulleyn L, Reardon W, et al: Identical mutations<br />

in the FGFR2 gene cause both Pfeiffer and Crouzon<br />

syndrome phenotypes. Nature Genet 9:173, 1995.<br />

183. Wilkie A, Slaney S, Oldridge M, et al: Apert syndrome<br />

results from localized mutations of FGFR2 and is allelic with<br />

Crouzon syndrome. Nature Genet 9:165, 1995.<br />

184. Lajeunie E, Ma H, Bonaventure J, et al: FGFR2 mutations<br />

in Pfeiffer syndrome (letter). Nature Genet 9:108, 1995.<br />

185. Gripp KW, McDonald-McGinn D, Gaudenz K, et al:<br />

Identification of a genetic cause for isolated unilateral<br />

coronal synostosis: a unique mutation in the fibroblast<br />

growth factor receptor 3. J Pediatr 132:714, 1998.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

186. Mulliken JB, Steinberger D, Kunze S, Muller U: Molecular<br />

diagnosis of bilateral coronal synostosis. Plast Reconstr<br />

Surg 104:1603, 1999.<br />

187. Biesecker LG: Strike three for GLI3. Nat Genet 17:259,<br />

1997.<br />

188. Vortkamp A, Gessler M, Grzeschik KH: GLI3 zinc-finger<br />

gene interrupted by translocations in Greig syndrome<br />

families. Nature 352:539, 1991.<br />

189. Wild A, Kalff-Suske M, Vortkamp A, et al: Point mutations<br />

in human GLI3 cause Greig syndrome. Hum Mol Genet<br />

6:1979, 1997.<br />

190. Lee MS, Lowe GN, Strong DD, et al: TWIST, a basic helixloop-helix<br />

transcription factor, can regulate the human<br />

osteogenic lineage. J Cell Biochem 75:566, 1999.<br />

191. Ghouzzi V el, Le Merrer M, Perrin-Schmitt F, et al: Mutations<br />

of the TWIST gene in the Saethre-Chotzen syndrome.<br />

Nat Genet 15:42, 1997.<br />

192. Howard TD, Paznekas WA, Green ED, et al: Mutations in<br />

TWIST, a basic helix-loop-helix transcription factor, in<br />

Saethre-Chotzen syndrome. Nat Genet 15:36, 1997.<br />

193. Paznekas WA, Cunningham ML, Howard TD, et al: Genetic<br />

heterogeneity of Saethre-Chotzen syndrome, due to<br />

TWIST and FGFR mutations. Am J Hum Genet 62:1370,<br />

1998.<br />

194. Rice DP, Aberg T, Chan Y, et al: Integration of FGF and<br />

TWIST in calvarial bone and suture development. Development<br />

127:1845, 2000.<br />

195. Ting K, Vastardis H, Mulliken JB, et al: Human NELL-1<br />

expressed in unilateral coronal synostosis. J Bone Miner Res<br />

14:80, 1999.<br />

196. Ting K: Personal communication, 2000. Cited in Warren<br />

SM, Greenwald JA, Spector JA, et al: New developments<br />

in cranial suture research. Plast Reconstr Surg 107:523,<br />

2001.<br />

197. Warren SM, Greenwald JA, Spector JA, et al: New developments<br />

in cranial suture research. Plast Reconstr Surg<br />

107:523, 2001.<br />

198. Kaiser G: Sagittal synostosis—Clinical significance and<br />

results of three different methods of craniectomy. Child’s<br />

Nerv Syst 4:223, 1988.<br />

199. Marchac D, Renier D: Intracranial pressure in craniosynostosis.<br />

J Neurosurg 57:370, 1982.<br />

200. Renier D, Marchac D: <strong>Craniofacial</strong> surgery for craniosynostosis:<br />

Functional and morphological results. Ann<br />

Acad Med Singapore 17:415, 1988.<br />

201. Fok H et al: Relationship between intracranial pressure<br />

and intracranial volume in craniosynostosis. Br J Plast Surg<br />

45:394, 1992.<br />

202. Gault DT et al: Intracranial pressure and intracranial<br />

volume in children with craniosynostosis. Plast Reconstr<br />

Surg 90:377, 1992.<br />

203. Kapp-Simon KA et al: Longitudinal assessment of mental<br />

development in infants with nonsyndromic craniosynostosis<br />

with and without cranial release and reconstruction.<br />

Plast Reconstr Surg 92:831, 1993.<br />

204. Renier D, Marchac D: Discussion of “Longitudinal assessment<br />

of mental development in infants with nonsyndromic<br />

craniosynostosis with and without cranial release and reconstruction,”<br />

by K A Kapp-Simon et al. Plast Reconstr<br />

Surg 92:840, 1993.<br />

205. Kapp-Simon KA: Mental development and learning disorders<br />

in children with single suture craniosynostosis. Cleft<br />

Palate Craniofac J 35:197, 1998.<br />

55


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

206. Virtanen R, Korhonen T, Fagerholm J, Viljanto J:<br />

Neurocognitive sequelae of scaphocephaly. Pediatrics<br />

103(4 Pt 1):791, 1999.<br />

207. Gault DT et al: Intracranial volume in children with<br />

craniosynostosis. J Craniofac Surg 1:1, 1990.<br />

208. Gosain AK: Discussion of Polley JW, Charbel FT, Kim D,<br />

MaFee MF: Nonsyndromal craniosynostosis: longitudinal<br />

outcome following cranio-orbital reconstruction in infancy.<br />

Plast Reconstr Surg 102:629, 1998.<br />

209. Posnick JC et al: Indirect intracranial volume measurements<br />

using CT scans: Clinical applications for craniosynostosis.<br />

Plast Reconstr Surg 89:34, 1992.<br />

210. Polley JW, Charbel FT, Kim D, MaFee MF: Nonsyndromal<br />

craniosynostosis: longitudinal outcome following cranioorbital<br />

reconstruction in infancy. Plast Reconstr Surg<br />

102:619, 1998.<br />

211. David LR, Wilson JA, Watson NE, Argenta LC: Cerebral<br />

perfusion defects secondary to simple craniosynostosis. J<br />

Craniofac Surg 7:177, 1996.<br />

212. Taylor WJ, Hayward RD, Lasjaunias P, et al: Enigma of<br />

raised intracranial pressure in patients with complex craniosynostosis:<br />

the role of abnormal intracranial venous<br />

drainage. J Neurosurg 94:377, 2001.<br />

213. Cohen SR, Persing JA: Intracranial pressure in single-suture<br />

craniosynostosis. Cleft Palate Craniofac J 35:194, 1998.<br />

214. Barritt J, Brooksbank M, Simpson D: Scaphocephaly:<br />

Aesthetic and psychosocial considerations. Dev Med Child<br />

Neurol 23:183, 1981.<br />

215. Arndt EM et al: Psychosocial adjustment of 20 patients with<br />

Treacher Collins syndrome before and after reconstructive<br />

surgery. Br J Plast Surg 40:605, 1987.<br />

216. Pertschuk MJ, Whitaker LA: Psychosocial outcome of<br />

craniofacial surgery in children. Plast Reconstr Surg 82:741,<br />

1988.<br />

217. Ousterhout DK, Vargervik K: Aesthetic improvement<br />

resulting from craniofacial surgery in craniosynostosis syndromes.<br />

J Craniomaxillofac Surg 15:189, 1987.<br />

218. Barden RC et al: The physical attractiveness of facially<br />

deformed patients before and after craniofacial surgery.<br />

Plast Reconstr Surg 82:229, 1988.<br />

219. Barden RC et al: Emotional and behavioral reactions to<br />

facially deformed patients before and after craniofacial<br />

surgery. Plast Reconstr Surg 82:409, 1988.<br />

220. Collmann H, Sorensen N, Krauss J: Consensus: trigonocephaly.<br />

Childs Nerv Syst 12:664, 1996.<br />

221. Lajeunie E, Le Merrer M, Marchac D, Renier D:<br />

Syndromal and nonsyndromal primary trigonocephaly:<br />

analysis of a series of 237 patients. Am J Med Genet<br />

75:211, 1998.<br />

222. Sadove AM, Kalsbeck JE, Eppley BL, Javed T: Modifications<br />

in the surgical correction of trigonocephaly. Plast Reconstr<br />

Surg 85:853, 1990.<br />

223. Thompson DN, Malcolm GP, Jones BM, et al: Intracranial<br />

pressure in single-suture craniosynostosis. Pediatr Neurosurg<br />

22:235, 1995.<br />

224. Sidoti EJ Jr, Marsh JL, Marty-Grames L, Noetzel MJ: Longterm<br />

studies of metopic synostosis: frequency of cognitive<br />

impairment and behavioral disturbances. Plast Reconstr<br />

Surg 97:276, 1996.<br />

225. Bottero L, Lajeunie E, Arnaud E, et al: Functional outcome<br />

after surgery for trigonocephaly. Plast Reconstr Surg<br />

102:952, 1998.<br />

56<br />

226. Posnick JC, Lin KY, Chen P, Armstrong D: Metopic<br />

synostosis: quantitative assessment of presenting deformity<br />

and surgical results based on CT scans. Plast Reconstr<br />

Surg 93:16, 1997.<br />

227. Havlik RJ, Azurin DJ, Bartlett SP, Whitaker LA: Analysis and<br />

treatment of severe trigonocephaly. Plast Reconstr Surg<br />

103:381, 1999.<br />

228. McCarthy JG, Glasberg SB, Cutting CB, et al: Twenty-year<br />

experience with early surgery for craniosynostosis: I.<br />

Isolated craniofacial synostosis—results and unsolved problems.<br />

Plast Reconstr Surg 96:272, 1995.<br />

229. McCarthy JG, Karp NS, LaTrenta GS, Thorne GH: The<br />

effect of early fronto-orbital advancement on frontal sinus<br />

development and forehead aesthetics. Plast Reconstr Surg<br />

86:1078, 1990.<br />

230. Anderson H, Paranchos GS: Craniosynostosis: a review of<br />

the literature and indications for surgery. Acta Pediatr<br />

Scand 57:47, 1968.<br />

231. Burstein FD, Hudgins RJ, Cohen SR, Boydston WR: Surgical<br />

correction of severe scaphocephalic deformities. J<br />

Craniofac Surg 5(4):228, 1994.<br />

232. Persing JA, Jane JA, Edgerton MT: Surgical treatment of<br />

craniosynostosis. In: Persing JA, Edgerton MT, Jane JA (eds),<br />

Scientific Foundations of Surgical Treatment of Craniosynostosis.<br />

Baltimore, Williams & Wilkins, 1989, p 117.<br />

233. Ingraham FD, Alexander E Jr, Matson DD: Clinical studies<br />

in craniosynostosis: analysis of fifty cases and description of<br />

a method of surgical treatment. <strong>Surgery</strong> 27:518, 1948.<br />

234. Venes JL, Sayers MP: Sagittal synostectomy. Technical<br />

note. J Neurosurg 44:390, 1976.<br />

235. Olds MV, Storrs B, Walker ML: Surgical treatment of sagittal<br />

synostosis. Neurosurgery 18:345, 1986.<br />

236. Greene CS Jr, Winston KR: Treatment of scaphocephaly<br />

with sagittal craniectomy and biparietal morcellation.<br />

Neurosurgery 23:196, 1988.<br />

237. Ocampo RV Jr, Persing JA: Sagittal synostosis. Clin Plast<br />

Surg 21(4):563, 1994.<br />

238. Esparza J, Cordobes F, Munoz MJ, et al: Tratamiento de la<br />

craneosinostosis sagital (escaphocefalia), por medio de la<br />

correccion quirurgica inmediata. An Esp Pediatr 45:143,<br />

1996.<br />

239. Friede H, Lauritzen C, Figueroa AA: Roentgencephalometric<br />

follow-up after early osteotomies in patients<br />

with scaphocephaly. J Craniofac Surg 7:96, 1996.<br />

240. Norwood CW, Alexander E, Davis CH, Kelly DL: Recurrent<br />

and multiple suture closures after craniectomy for<br />

craniosynostosis. J Neurosurg 41:715, 1974.<br />

241. Jane JA, Edgerton MT, Futrell JW, Park TS: Immediate<br />

correction of sagittal synostosis. J Neurosurg 49:705, 1978.<br />

242. Vollmer DG, Jane JA, Park TS, Persing JA: Variants of sagittal<br />

synostosis: strategies for surgical correction. J Neurosurg<br />

61:557, 1984.<br />

243. Chadduck WM, Chadduck JB, Boop FA: The subarachnoid<br />

spaces in craniosynostosis. Neurosurgery 30:867, 1992.<br />

244. Epstein N, Epstein F, Newman G: Total vertex craniectomy for<br />

the treatment of scaphocephaly. Childs Brain 9:309, 1982.<br />

245. Marsh JL, Jenny A, Galic M, et al: Surgical management of<br />

sagittal synostosis. A quantitative evaluation of two techniques.<br />

Neurosurg Clin North Am 3:629, 1991.<br />

246. Christophis P, Junger TH, Howaldt H-P: Surgical correction of<br />

scaphocephaly: experiences with a new procedure and<br />

follow-up investigations. J Craniomaxillofac Surg 29:33, 2001.


247. Sutton LN, Barlett SP, Duhaine AC, Markakis D: Total<br />

cranial vault reconstruction for the older child with scaphocephaly.<br />

Pediatr Neurosurg 19:63, 1993.<br />

248. Alvarez-Garijo JA, Cavadas PC, Vila MM, Alvarez-Llanas A:<br />

Sagittal synostosis: results of surgical treatment in 210<br />

patients. Childs Nerv Syst 17:64, 2001.<br />

249. Marchac D, Renier D, Broumand S: Timing of treatment<br />

for craniosynostosis and faciocraniosynostosis: a 20-year<br />

experience. Br J Plast Surg 47:211, 1994.<br />

250. Snively SL, Albin M: Sagittal synostosis in the older patient.<br />

Presented at the Annual Meeting of the American Cleft<br />

Palate Association, San Francisco, April 1989.<br />

251. Mulliken JB, Vander Woude DL, Hansen M, et al: Analysis<br />

of posterior plagiocephaly: deformational versus synostotic.<br />

Plast Reconstr Surg 103:371, 1999.<br />

252. Huang MHS, Mouradian WE, Cohen SR, Gruss JS: The<br />

differential diagnosis of abnormal head shapes: separating<br />

craniosynostosis from positional deformities and normal<br />

variants. Cleft Palate Craniofac J 35:204, 1998.<br />

253. McComb J: Treatment of functional lambdoid synostosis.<br />

Neurosurg Clin North Am 2:665, 1991.<br />

254. O’Broin ES, Allcutt D, Earley MJ: Posterior plagiocephaly:<br />

proactive conservative management. Br J Plast Surg 52:18,<br />

1999.<br />

255. Ellenbogen RG, Gruss JS, Cunningham ML: Update on<br />

craniofacial surgery: the differential diagnosis of lambdoid<br />

synostosis/posterior plagiocephaly. Clin Neurosurg 47:303,<br />

2000.<br />

256. David DJ, Menard RM: Occipital plagiocephaly. Br J Plast<br />

Surg 53:367, 2000.<br />

257. Persing JA et al: Lambdoid synostosis: Surgical considerations.<br />

Plast Reconstr Surg 81:852, 1988.<br />

258. Mohr G, Hoffman HJ, Munro IR, et al: Surgical management<br />

of unilateral and bilateral coronal craniosynostosis:<br />

21 years of experience. Neurosurgery 2:83, 1978.<br />

259. Hansen M, Mulliken JB: Frontal plagiocephaly. Diagnosis<br />

and treatment. Clin Plast Surg 21(4):543, 1994.<br />

260. Marsh JL, Gado MH, Vannier MW, Stevens WG: Osseous<br />

anatomy of unilateral coronal synostosis. Cleft Palate J<br />

23:87, 1986.<br />

261. Bruneteau RJ, Mulliken JB: Frontal plagiocephaly: synostotic,<br />

compensational, or deformational. Plast Reconstr Surg<br />

89:21, 1992.<br />

262. Bruneteau RJ, Mulliken JB: Frontal plagiocephaly: Synostotic,<br />

compensational, or deformational. Plast Reconstr<br />

Surg 89:21, 1992.<br />

263. Lo LJ, Marsh JL, Pilgram TK, Vannier MW: Plagiocephaly:<br />

differential diagnosis based on endocranial dysmorphology.<br />

Plast Reconstr Surg 97:282, 1996.<br />

264. Lo L-J, Marsh JL, Kane AA, Vannier MW: Orbital<br />

dysmorphology in unilateral coronal synostosis. Cleft Palate<br />

Craniofac J 33:190, 1996.<br />

265. Posnick JC: Anterior plagiocephaly: unilateral coronal<br />

synostosis and skull molding. In: Posnick JC (ed), <strong>Craniofacial</strong><br />

and Maxillofacial <strong>Surgery</strong> in Children and Young<br />

Adults. Philadelphia, WB Saunders, 2000. Vol 1, Ch 9.<br />

266. Wagner JD, Cohen SR, Maher H, et al: Critical analysis of<br />

results of craniofacial surgery for nonsyndromic bicoronal<br />

synostosis. J Craniofac Surg 6(1):32, 1995.<br />

267. Bertelson TI: The premature synostosis of the cranial<br />

sutures. Acta Ophthalmol 51:1, 1958.<br />

268. Marchac D, Renier D, Jones BM: Experience with the<br />

“floating forehead.” Br J Plast Surg 41:1, 1988.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

269. McCarthy JG, Glasberg SB, Cutting CB, et al: Twenty-year<br />

experience with early surgery for craniosynostosis: II. The<br />

craniofacial synostosis syndromes and pansynostosis—results<br />

and unsolved problems. Plast Reconstr Surg 96:284,<br />

1995.<br />

270. Persing J, Babler W, Winn HR, et al: Age as a critical factor<br />

in the success of surgical correction of craniosynostosis. J<br />

Neurosurg 54:601, 1981.<br />

271. Cohen MM Jr: Phenotypic/molecular correlations. Am J<br />

Med Genet 56:334, 1995.<br />

272. Wheaton SW: Two specimens of congenital cranial<br />

deformities in infants in association with fusion of the<br />

fingers and toes. In: Smith DW (ed), Recognizable<br />

Patterns of Human malformation. Major Problems in<br />

Clinical Pediatrics Vol 7, Philadelphia, WB Saunders,<br />

1982, p 308.<br />

273. Tessier P: Apert’s Syndrome: Acrocephalosyndactyly<br />

Type I. In: Caronni EP (ed), <strong>Craniofacial</strong> <strong>Surgery</strong>. Boston,<br />

Little Brown, 1985. Ch 27, pp 280-303.<br />

274. Jackson IT: <strong>Craniofacial</strong> Dysostosis. In: Serafin D and<br />

Georgiade NG (eds), Pediatric <strong>Plastic</strong> <strong>Surgery</strong>. St Louis,<br />

CV Mosby, 1984. Vol 1, Ch 27, pp 440-466.<br />

275. Tessier P: Relationship of craniostenoses to craniofacial<br />

dysostoses and to faciostenoses. A study with therapeutic<br />

implications. Plast Reconstr Surg 48:224, 1971.<br />

276. Pollard ZF: Bilateral superior oblique muscle palsy associated<br />

with Apert’s syndrome. Am J Ophthalmol 106:337,<br />

1988.<br />

277. Ousterhout DK, Melsen B: Cranial base deformity in<br />

Apert’s syndrome. Plast Reconstr Surg 69:254, 1982.<br />

278. Upton J: Apert syndrome. Classification and pathologic<br />

anatomy of limb anomalies. Clin Plast Surg 18(2):321,<br />

1991.<br />

279. Clauser L, Galie M, Hassanipour A, Calabrese O: Saethre-<br />

Chotzen syndrome: review of the literature and report of<br />

a case. J Craniofac Surg 11:480, 2000.<br />

280. Cohen MM Jr: Pfeiffer syndrome update: clinical subtypes<br />

and guidelines for differential diagnosis. Am J Med Genet<br />

45:300, 1993.<br />

281. Moore MH, Lodge ML, Clark BE: The infant skull in<br />

Pfeiffer’s syndrome. J Craniofac Surg 6(6):483, 1995.<br />

282. Winter RM, Pfeiffer syndrome. Am J Med Genet 49(3):357,<br />

1994.<br />

283. Muenke M, Schell U, Hehr A, et al: A common mutation<br />

in the fibroblast growth factor receptor 1 gene in Pfeiffer<br />

syndrome. Nature Genet 8:269, 1994.<br />

284. Kerr NC, Wilroy RS Jr, Kaufman RA: Type 3 Pfeiffer<br />

syndrome with normal thumbs. Am J Med Genet 66:138,<br />

1996.<br />

285. Schell U, Hehr A, Feldman GH, et al: Mutations in FGFR1<br />

and FGFR2 cause familial and sporadic Pfeiffer syndrome<br />

Hum Mol Genet 4:323, 1995.<br />

286. Taravath S, Tonsgard JH: Cerebral malformations in Carpenter<br />

syndrome. Pediatr Neurol 9:230, 1993.<br />

287. Poole MD: Surgical caution with Carpenter’s syndrome. J<br />

Craniomaxillofac Surg 21:93, 1993.<br />

288. Tessier P: The definitive plastic surgical treatment of the<br />

severe facial deformities of craniofacial dysostosis. Crouzon’s<br />

and Apert’s diseases. Plast Reconstr Surg 48:419, 1971.<br />

289. Cohen MM Jr: Craniosynostosis and syndromes with<br />

craniosynostosis: incidence, genetics, penetrance, variability,<br />

and new syndrome updating. Birth Defects<br />

15(5B):13, 1979.<br />

57


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

290. Dodge HW, Wood MW, Kennedy RL: <strong>Craniofacial</strong> dysostosis:<br />

Crouzon’s disease. J Pediatr 23:98, 1959.<br />

291. Costaras-Volarich M, Pruzansky S: Is the mandible intrinsically<br />

different in Apert and Crouzon syndromes? Am J<br />

Orthod 85:475, 1984.<br />

292. Rosen HM, Whitaker LA: Cranial base dynamics in craniofacial<br />

dysostosis. J Maxillofac Surg 12:56, 1984.<br />

293. Proudman TW, Moore MH, Abbott AH, David DJ:<br />

Noncraniofacial manifestations of Crouzon’s disease. J<br />

Craniofac Surg 5(4):218, 1994.<br />

294. Kreiborg S: Crouzon syndrome: A clinical and<br />

roentgencephalometric study. Scand J Plast Reconstr Surg<br />

18 (Suppl):38, 1981.<br />

295. Kreiborg S et al: Comparative three-dimensional analysis<br />

of CT-scans of the calvaria and cranial base in Apert and<br />

Crouzon syndromes. J Craniomaxillofac Surg 21:181,<br />

1993.<br />

296. Posnick JC, Liin KY, Khawar BJ, Armstrong D: Crouzon<br />

syndrome: quantitative assessment of presenting deformity<br />

and surgical results based on CT scans. Plast Reconstr<br />

Surg 92:1027, 1993.<br />

297. Cohen MMJ: Craniofrontonasal dysplasia. Birth Defects<br />

15:85, 1979.<br />

298. Sedano HE, Gorlin RJ: Frontonasal malformation as a field<br />

defect and in syndromic associations. Oral Surg 65:704,<br />

1988.<br />

299. Orr DJA, Slaney S, Ashworth GJ, Poole MD: Cranio<br />

frontonasal dysplasia. Br J Plast Surg 50:153, 1997.<br />

300. Hemmer KM, McAlister WH, Marsh JL: Cervical spine<br />

anomalies in the craniosynostosis syndromes. Cleft Palate<br />

J 24:328, 1987.<br />

301. Parry CH: Collections from the Unpublished Medical<br />

Writings of the Late Caleb Hillier Parry. London,<br />

Underwoods, 1825, p 478-480.<br />

302. Romberg MH: Klinische Ergebnisse. Berlin, A Forstner,<br />

1846, p 75-81.<br />

303. Eulenberg A: Lehrbuch der functionellen Nervenkrankheiten.<br />

Berlin, S Karger, 1871.<br />

304. Rogers BO: Progressive facial hemiatrophy (Romberg’s<br />

disease): a review of 772 cases. In: Transactions of the<br />

Third International Congress of <strong>Plastic</strong> <strong>Surgery</strong>. Washington,<br />

Excerpta Medica, 1963, p 681-689.<br />

305. Moebius PJ: Der umschriebene Gesichtsschwund. In:<br />

Northanagel CWH (ed), Specielle Pathologie und<br />

Therapie, Vol 2, Pt 2. Vienna, A Holder, 1912.<br />

306. Mendel G: Uber Hemiatrophia facialis. Dtsch Med<br />

Wochenschr 14:321, 1888.<br />

307. Moss ML, Crikelair GJ: Progressive facial hemiatrophy<br />

following cervical sympathectomy in the rat. Arch Oral Biol<br />

1:254, 1960.<br />

308. Pensler JM, Murphy GF, Mulliken JB: Clinical and ultrastructural<br />

studies of Romberg’s hemifacial atrophy. Plast<br />

Reconstr Surg 85:669, 1990.<br />

309. Moore MH, Wong KS, Proudman TW, David DJ: Progressive<br />

hemifacial atrophy (Romberg’s disease): skeletal<br />

involvement and treatment. Br J Plast Surg 46:39,<br />

1993.<br />

310. Jurkiewicz MJ, Nahai F: The use of free revascularized<br />

grafts in the amelioration of hemifacial atrophy. Plast<br />

Reconstr Surg 76:44, 1985.<br />

311. Inigo F, Rojo P, Ysunza A: Aesthetic treatment of Romberg’s<br />

disease: experience with 35 cases. Br J Plast Surg 46:194,<br />

1993.<br />

58<br />

312. Dunkley MP, Stevenson JH: Experience with the free<br />

“inverted” groin flap in facial soft tissue contouring; a<br />

report on 6 flaps. Br J Plast Surg 43:154, 1990.<br />

313. Tweed AEJ, Manktelow RT, Zuker RM: Facial contour<br />

reconstruction with free flaps. Ann Plast Surg 12:313,<br />

1984.<br />

314. Harashina T, Fujino T: Reconstruction in Romberg’s<br />

disease with free groin flap. Ann Plast Surg 7:289, 1981.<br />

315. Williams HB, Crepeau RJ: Free dermal fat flaps to the face.<br />

Ann Plast Surg 3:1, 1979.<br />

316. Koshima I, Inagawa K, Urushibara K, et al: Deep inferior<br />

epigastric perforator dermal-fat or adiposal flap for correction<br />

of craniofacial contour deformities. Plast Reconstr<br />

Surg 106:10, 2000.<br />

317. Mordick TG II, Larossa D, Whitaker L: Soft-tissue reconstruction<br />

of the face: a comparison of dermal-fat grafting<br />

and vascularized tissue transfer. Ann Plast Surg 29:390,<br />

1992.<br />

318. Upton J, Albin RE, Mulliken JB, Murray JE: The use of<br />

scapular and parascapular flaps for cheek reconstruction.<br />

Plast Reconstr Surg 90:959, 1992.<br />

319. Longaker MT, Siebert JW: Microvascular free-flap correction<br />

of severe hemifacial atrophy. Plast Reconstr Surg<br />

96:800, 1995.<br />

320. Rees TD, Ashley FL, Delgado JP: Silicone fluid injections<br />

for facial atrophy: a 10-year study. Plast Reconstr Surg<br />

52:118, 1973.<br />

321. De la Fuente A, Tavora T: fat injections for the correction<br />

of facial lipodistrophies: a preliminary report. Aesthetic<br />

Plast Surg 12:39, 1988.<br />

322. De la Fuente A, Jimenez A: Latissimus dorsi free flaps for<br />

restoration of facial contour defects. Ann Plast Surg 22:1,<br />

1989.<br />

323. Jones GC et al: Juvenile neurofibroma: Behavior and<br />

treatment of extensive and residual tumors. Arch<br />

Otolaryngol 112:1191, 1986.<br />

324. Nichols RD et al: Meningioma in the parotid region.<br />

Laryngoscope 97:693, 1987.<br />

325. Humphreys DH, Schwartz MR, Jenkins HA: Meningioma:<br />

A case of transcranial recurrence managed by base-of-skull<br />

technique and a review of the tumor. Otolaryngol Head<br />

Neck Surg 93:563, 1985.<br />

326. Jackson IT: Management of <strong>Craniofacial</strong> Tumors. In:<br />

Caronni EP (ed), <strong>Craniofacial</strong> <strong>Surgery</strong>. Boston, Little<br />

Brown, 1982. Ch 29, pp 315-328.<br />

327. Shah JP et al: <strong>Craniofacial</strong> resections for tumors involving<br />

the base of the skull. Am J Surg 154:352, 1987.<br />

328. Kennerdell JS, Maroon JC, Malton ML: Surgical approaches<br />

to orbital tumors. Clin Plast Surg 15:273, 1988.<br />

329. Sundaresan N, Shah JP: <strong>Craniofacial</strong> resection for anterior<br />

skull base tumors. Head Neck Surg 10:219, 1988.<br />

330. Panje WR et al: The transfacial approach for combined<br />

anterior craniofacial tumor ablation. Arch Otolaryngol<br />

115:301, 1989.<br />

331. Jackson IT, Adham MN, Marsh WR: Use of the galeal<br />

frontalis myofascial flap in craniofacial surgery. Plast Reconstr<br />

Surg 77:905, 1986.<br />

332. Arden RL, Mathog RH, Thomas LM: Temporalis musclegalea<br />

flap in craniofacial reconstruction. Laryngoscope<br />

97:1336, 1987.<br />

333. Fisher J, Jackson IT: Microvascular surgery as an adjunct to<br />

craniomaxillofacial reconstruction. Br J Plast Surg 42:146,<br />

1989.


334. Colen SR: Selective microvascular procedures in<br />

oculoplastic surgery. Clin Plast Surg 15:283, 1988.<br />

335. Cohen SR, Bartlett SP, Whitaker LA: Reconstruction of late<br />

craniofacial deformities after irradiation of the head and<br />

face during childhood. Plast Reconstr Surg 86:229, 1990.<br />

336. Edgerton MT, Persing JA, Jane JA: The surgical treatment<br />

of fibrous dysplasia. With emphasis on recent contributions<br />

from craniomaxillofacial surgery. Ann Surg 202:459,<br />

1985.<br />

337. Ricalde P, Horswell BB: <strong>Craniofacial</strong> fibrous dysplasia of<br />

the fronto-orbital region: a case series and literature<br />

review. J Oral Maxillofac Surg 59:157, 2001.<br />

338. Barat M, Rybak LP, Mann JL: Fibrous dysplasia masquerading<br />

as chronic maxillary sinusitis. Ear Nose Throat J 68:42,<br />

1989.<br />

339. Posnick JC: Fibrous dysplasia of the craniomaxillofacial<br />

region: current clinical perspectives. Br J Oral Maxillofac<br />

Surg 36:264, 1998.<br />

340. Liakos GM, Walker CB, Carruth JAS: Ocular complications<br />

in fibrous dysplasia. Br J Ophthalmol 68:611, 1979.<br />

341. Forbes G, Gorman CA, Brennan MD, et al: Ophthalmalogy<br />

of Graves disease: Computerized volume measurements<br />

of the orbital fat in muscles. Am J Neuroradiol 7(4):651,<br />

1986.<br />

342. Lello GE amd Sparrow OC: <strong>Craniofacial</strong> polyostotic fibrous<br />

dysplasia. J Maxillofac Surg 13:267, 1985.<br />

343. Papay F, Morales L, Flaharty P, et al: Optic nerve decompression<br />

in cranial base fibrous dysplasia. J Craniofac Surg<br />

6(1):5, 1995.<br />

344. Schwartz DT, Alpert M: The malignant transformation of<br />

fibrous dysplasia. Am J Med 32:393, 1962.<br />

345. Zachariades N et al: Cherubism. Int J Oral Surg 14:138,<br />

1985.<br />

346. Van Horn PE Jr, Dahlin DC, Bickel WH: Fibrous Dysplasia:<br />

a clinical study of orthopedic surgical cases. Mayo Clin<br />

Proc 38:175, 1963.<br />

347. Derome P: Les tumeurs spheno-ethmodiales (possibilites<br />

d’exenese et de reparation chirurgicales). Rapport de la<br />

Societe de Neuro- chirurgie de Langue Francaise.<br />

348. Chen Y-R, Breidahl A, Chang C-N: Optic nerve decompression<br />

in fibrous dysplasia: indications, efficacy, and<br />

safety. Plast Reconstr Surg 99:22, 1997.<br />

349. Weisman JS, Hepler RS, Vinters HV: Reversible visual loss<br />

caused by fibrous dysplasia. Am J Ophthalmol 110:244,<br />

1990.<br />

350. Lauritzen C, Alberius P, Santanelli F, et al: Repositioning of<br />

craniofacial tumorous bone after autoclaving. Scand J Plast<br />

Reconstr Hand Surg 25:161, 1991.<br />

351. Bradley PF: A 2-stage procedure for reimplantation of<br />

autogenous freeze-treated mandibular bone. J Oral<br />

Maxillofac Surg 40:278, 1982.<br />

352. Bradley PF: Modern trends in cryosurgery of bone in the<br />

maxillo-facial region. Int J Oral Surg 7:405, 1978.<br />

353. Chen Y-R, Noordhoff MS: Treatment of craniomaxillofacial<br />

fibrous dysplasia: How early and how extensive? Plast<br />

Reconstr Surg 86:835, 1990.<br />

354. Hansen-Knarhoi M, Poole MD: Preoperative difficulties in<br />

differentiating intraosseous meningiomas and fibrous dysplasia<br />

around the orbital apex. J Craniomaxillofac Surg<br />

22(4):226, 1994.<br />

355. Chapurlat RD, Delmas PD, Liens D, Meunier PJ: Longterm<br />

effects of intravenous pamidronate in fibrous dysplasia<br />

of bone. J Bone Miner Res 12:1746, 1997.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

356. Clark C, Hobar PC, and Rajagopal U: Characterization of<br />

fibrous dysplasia in the nude mouse model: Evidence for<br />

estrogen dependent growth. Surgical Forum 44:741,<br />

1993.<br />

357. Krastinova-Lolov K, Hamza F: the surgical management of<br />

cranio-orbital neurofibromatosis. Ann Plast Surg 36(3):263,<br />

1996.<br />

358. Poole MD: Experiences in the surgical treatment of<br />

cranioorbital neurofibromatosis. Br J Plast Surg 42:155,<br />

1989.<br />

359. Marchac D: Intracranial enlargement of the orbital cavity<br />

and palpebral remodeling for orbitopalpebral neurofibromatosis.<br />

Plast Reconstr Surg 73:534, 1984.<br />

360. Snyder BJ, Hanieh A, Trott JA, David DJ: Transcranial<br />

correction of orbital neurofibromatosis. Plast Reconstr<br />

Surg 102:633, 1998.<br />

361. Jackson IT et al: Orbitotemporal neurofibromatosis: Classification<br />

and treatment. Plast Reconstr Surg 92:1, 1993.<br />

362. Lannelongue M: De la craniectomie dans la microcephalie.<br />

C R Acad Sci Paris 110:1382, 1890.<br />

363. Lane LC: Pioneer craniectomy for relief of mental imbecility<br />

due to premature sutural closure and microcephalus.<br />

JAMA 18:49, 1892.<br />

364. Waterhouse N: The history of craniofacial surgery. Fac<br />

Plast Surg 9:143, 1993.<br />

365. Gillies H, Harrison SH: Operative correction by osteotomy<br />

of recessed malar maxillary compound in a case of oxycephaly.<br />

Br J Plast Surg 3:123, 1950-51.<br />

366. Converse JM, Smith B: An Operation for Congenital and<br />

Traumatic Hypertelorism. In: Troutman RC, Converse JM,<br />

Smith B (eds), <strong>Plastic</strong> and Reconstructive <strong>Surgery</strong> of the<br />

Eye and Adnexa. Washington, Butterworths, 1962.<br />

367. Tessier P: Osteotomies totales de la face. Syndrome de<br />

Crouzon. Syndrome d’Apert. Oxycephalies. Scaphocephalies.<br />

Turricephalies. Ann Chir Plast 12:273, 1967.<br />

368. Tessier P: Orbital hypertelorism: I. Successive surgical<br />

attempts. Materials and methods. Causes and mechanisms.<br />

Scand J Plast Reconstr Surg 6:135, 1972.<br />

369. Tessier P, Guiot G, Derome P: Orbital hypertelorism: II.<br />

Definite treatment of orbital hypertelorism (ORH) by craniofacial<br />

or by extracranial osteotomies. Scand J Plast<br />

Reconstr Surg 7:39, 1973.<br />

370. Tessier P: Experiences in the treatment of orbital<br />

hypertelorism. Plast Reconstr Surg 53:1, 1974.<br />

371. Le Fort R: Experimental study of fractures of the upper jaw.<br />

<strong>Part</strong>s I and II. Reprinted in Plast Reconstr Surg 50:497,<br />

1972. <strong>Part</strong> III, Plast Reconstr Surg 50:600, 1972.<br />

372. Caronni EP: Preface. In: Caronni EP (ed), <strong>Craniofacial</strong><br />

<strong>Surgery</strong>. Boston, Little Brown, 1982.<br />

373. Obwegeser HL: Surgical correction of dish-face deformity.<br />

Plast Reconstr Surg 43:351, 1969.<br />

374. Munro IR: The Luhr fixation system for the craniofacial<br />

skeleton. Clin Plast Surg 16:41, 1989.<br />

375. Marsh JL: The use of the Wurtzberg system to facilitate<br />

fixation in facial osteotomies. Clin Plast Surg 16:49, 1989.<br />

376. Munro IR: Rigid fixation and facial asymmetry. Clin Plast<br />

Surg 16:187, 1989.<br />

377. Fuente del Campo A: Rigid fixation and osteotomy design<br />

in frontal orbital advancement osteotomies. Clin Plast Surg<br />

16:205, 1989.<br />

378. Beals SP, Munro IR: The use of miniplates in<br />

craniomaxillofacial surgery. Plast Reconstr Surg 79:33,<br />

1987.<br />

59


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

379. Munro IR, Fearon JA, Bruce DA: Complications of rigid<br />

fixation in infants and small children. Presented at the Fifth<br />

International Congress of the International Society of<br />

<strong>Craniofacial</strong> <strong>Surgery</strong>, Oaxaca, Mexico, October 1993.<br />

380. Yu JY, Goldberg DS, Bartlett SP, Whitaker LA: The long<br />

term positional stability of microfixation on the growing<br />

swing craniofacial skeleton. Presented at the annual meeting<br />

of the American Society of <strong>Plastic</strong> and Reconstructive<br />

Surgeons, San Diego, CA, September 1994.<br />

381. Goldberg DS, Bartlett SP, Yu JC, et al: Critical review of<br />

microfixation in pediatric craniofacial surgery. J Craniofac<br />

Surg 6(4):301, 1995.<br />

382. Papay FA, Hardy S, Morales L Jr, et al: “False” migration of<br />

rigid fixation appliances in pediatric craniofacial surgery. J<br />

Craniofac Surg 6(4):309, 1995.<br />

383. Berryhill WE, Rimell FL, Ness J, et al: Fate of rigid fixation<br />

in pediatric craniofacial surgery. Otolaryngol Head Neck<br />

Surg 121:269, 1999.<br />

384. Orringer JS, Barcelona V, Buchman SR: Reasons for<br />

removal of rigid internal fixation devices in craniofacial<br />

surgery. J Craniofac Surg 9:40, 1998.<br />

385. Ahn DK, Sims CD, Randolph MA, et al: <strong>Craniofacial</strong><br />

skeletal fixation using biodegradable plates and cyanoacrylate<br />

glue. Plast Reconstr Surg 99:1508, 1997.<br />

386. Eppley BL, Sadove AM: Effects of resorbable fixation on<br />

craniofacial skeletal growth: alteration in plate size. J<br />

Craniofac Surg 5:110, 1994.<br />

387. Eppley BL, Sadove AM: Resorbable coupling fixation in<br />

craniosynostosis surgery: experimental and clinical applications.<br />

J Craniofac Surg 6(6):477, 1995.<br />

388. Wiltfang J, Merten HA, Schultze-Mosgau S, et al: Biodegradable<br />

miniplates (LactoSorb): long-term results in infant<br />

minipigs and clinical results. J Craniofac Surg 11:239,<br />

2000.<br />

389. Tharanon W, Sinn DP, Hobar PC, et al: Surgical outcomes<br />

using bioabsorbable plating systems in pediatric craniofacial<br />

surgery. J Craniofac Surg 9:441, 1998.<br />

390. Kurpad SN, Goldstein JA, Cohen AR: Bioresorbable fixation<br />

for congenital pediatric craniofacial surgery: a 2-year<br />

follow-up. Pediatr Neurosurg 33:306, 2000.<br />

391. Imola MJ, Hamlar DD, Shao W, et al: Resorbable plate<br />

fixation in pediatric craniofacial surgery: long-term outcome.<br />

Arch Facial Plast Surg 3:79, 2001.<br />

392. Ambay RS, Mount DL: <strong>Craniofacial</strong> <strong>Anomalies</strong> I: Cephalometries<br />

and Orthognathic <strong>Surgery</strong>. Selected Read Plast<br />

Surg 10(17) Pt 1, 2007.<br />

393. Shillito J, Matson DD: Craniosynostosis: A review of 519<br />

surgical patients. Pediatrics 41:829, 1968.<br />

394. Epstein N, Epstein F, Newman G: Total vertex craniectomy<br />

for the treatment of scaphocephaly. Child’s Brain 9:309,<br />

1982.<br />

395. Hoffmann HJ, Mohr G: Lateral canthal advancement of the<br />

supraorbital margins. J Neurosurg 45:376, 1976.<br />

396. Marchac D: Radical forehead remodeling for craniostenosis.<br />

Plast Reconstr Surg 61:823, 1978.<br />

397. Marchac D, Renier D: <strong>Craniofacial</strong> surgery for craniosynostosis.<br />

Scand J Plast Reconstr Surg 15:235, 1981.<br />

398. Hendel PM, Nadell JM: Projection geometry and stress–<br />

reduction techniques in craniofacial surgery. Plast Reconstr<br />

Surg 83:217, 1989.<br />

399. Albin RE et al: Trigonocephaly: Refinements in reconstruction.<br />

Experience with 33 patients. Plast Reconstr Surg<br />

76:202, 1985.<br />

60<br />

400. Munro IR, Hoffmann HJ, Hendrick EB: Total cranial vault<br />

reshaping in craniofacial surgery. In: Transactions of the<br />

Sixth International Congress of <strong>Plastic</strong> and Reconstructive<br />

<strong>Surgery</strong>, Paris, Masson, 1975, p 158.<br />

401. Jackson IT, Hide TAH, Barker DT: Transposition cranioplasty<br />

to restore forehead contour in craniofacial deformities.<br />

Br J Plast Surg 31:127, 1978.<br />

402. Ortiz Monasterio F, Fuente del Campo A, Carrillo A:<br />

Advancement of the orbits and the midface in one piece,<br />

combined with frontal repositioning for the correction of<br />

Crouzon’s deformities. Plast Reconstr Surg 61:507, 1978.<br />

403. Marchac D, Renier D: Commentary on cranial vault<br />

expansion. J Craniofac Surg 4:174, 1993.<br />

404. Marchac D, Renier D, Broumand S: Timing of treatment<br />

for craniosynostosis and faciocraniosynostosis: a 20-year<br />

experience. Br J Plast Surg 47:211, 1994.<br />

405. McCarthy JG et al: The effect of early fronto-orbital<br />

advancement on frontal sinus development and forehead<br />

aesthetics. Plast Reconstr Surg 86:1078, 1990.<br />

406. Bartlett SP, Whitaker LA, Marchac D: The operative<br />

treatment of isolated craniofacial dysostosis (plagiocephaly):<br />

A comparison of the unilateral and bilateral techniques.<br />

Plast Reconstr Surg 85:677, 1990.<br />

407. David DJ, Sheen R: Surgical correction of Crouzon syndrome.<br />

Plast Reconstr Surg 85:344, 1990.<br />

408. Marchac D, Renier D: Treatment of craniosynostosis in<br />

infancy. Clin Plast Surg 14:61, 1987.<br />

409. McCarthy JG et al: Early surgery for craniofacial synostosis—<br />

An 8-year experience. Plast Reconstr Surg 73:521, 1984.<br />

410. Dufresne CR et al: Volumetric quantification of intracranial<br />

and ventricular volume following cranial vault remodeling:<br />

A preliminary report. Plast Reconstr Surg 79:24, 1987.<br />

411. Marsh JL, Vannier MW: Cranial base changes following<br />

surgical treatment of craniosynostosis. Cleft Palate J 23(Suppl<br />

1):9, 1986.<br />

412. Friede H, Lilja J, Laurizen C, et al: Skull morphology after<br />

early craniotomy in patients with premature synostosis of<br />

the coronal suture. Cleft Palate J 23 (Suppl 1):1, 1986.<br />

413. Persing JA, Jane JA, Edgerton MT: Surgical Treatment of<br />

Craniosynostosis. In: Persing JA, Edgerton MT, Jane JA<br />

(eds), Scientific Foundations and Surgical Treatment of<br />

Craniosynostosis. Baltimore, Williams & Wilkins, 1989,<br />

pp 117-238.<br />

414. Heeckt P, Muhlbauer W, Anderl H, et al: Early radical<br />

treatment of pancraniofacial synostosis. Ann Plast Surg<br />

30:312, 1993.<br />

415. Whitaker LA et al: Craniosynostosis: An analysis of the<br />

timing, treatment, and complications in 164 consecutive<br />

patients. Plast Reconstr Surg 80:195, 1987.<br />

416. Roddi R, Vaandrager JM, Gilbert PM, van der Meulen JC:<br />

Reshaping of the skull in the early surgical correction of<br />

scaphocephaly. J Craniomaxillofac Surg 21:226, 1993.<br />

417. Wall SA, Goldin JH, Hockley AD, et al: Fronto-orbital reoperation<br />

in craniosynostosis. Br J Plast Surg 47:180, 1994.<br />

418. McCarthy JG, Glasberg SB, Cutting CB, et al: Twenty-year<br />

experience with early surgery for craniosynostosis: I.<br />

Isolated craniofacial synostosis—results and unsolved problems.<br />

Plast Reconstr Surg 96:272, 1995.<br />

419. McCarthy JG, Glasberg SB, Cutting CB, et al: Twenty-year<br />

experience with early surgery for craniosynostosis: II. The<br />

craniofacial synostosis syndromes and pansynostosis—results<br />

and unsolved problems. Plast Reconstr Surg 96:284,<br />

1995.


420. Tessier P: Dysostoses cranio-faciales. Osteotomies totales<br />

de la face. Transactions of the Fourth International<br />

Congress of <strong>Plastic</strong> and Reconstructive <strong>Surgery</strong>,<br />

Amsterdam, Excerpta Medica, 1969.<br />

421. Tessier P: Total osteotomy of the middle third of the face<br />

for faciostenosis or for sequelae of le Fort III fractures. Plast<br />

Reconstr Surg 48:533, 1971.<br />

422. Van der Meulen JC: The pursuit of symmetry in craniofacial<br />

surgery. Br J Plast Surg 29:85, 1976.<br />

423. Van der Meulen JC: Medial faciotomy. Br J Plast Surg<br />

32:339, 1979.<br />

424. Anderl H et al: Frontofacial advancement with bony<br />

separation in craniofacial dysostosis. Plast Reconstr Surg<br />

71:303, 1983.<br />

425. Wolfe SA et al: The monobloc frontofacial advancement:<br />

Do the pluses outweigh the minuses? Plast Reconstr Surg<br />

91:977, 1993.<br />

426. Fearon JA, Whitaker LA: Complications with facial advancement:<br />

A comparison between the Le Fort III and<br />

monobloc advancements. Plast Reconstr Surg 91:990,<br />

1993.<br />

427. Munro IR, Das SK: Improving results in orbital hypertelorism<br />

correction. Ann Plast Surg 2:499, 1979.<br />

428. Edgerton MT, Jane JA: Vertical orbital dystopia—Surgical<br />

correction. Plast Reconstr Surg 67:121, 1981.<br />

429. Byrd HS, Hobar PC: Optimizing the management of<br />

secondary zygomatic fracture deformities. Aesthetic and<br />

functional considerations. Clin Plast Surg 19:259, 1992.<br />

430. De Ponte FS, Fadda T, Rinna C, et al: Early and late surgical<br />

treatment of orbital dystopia in craniofacial malformation.<br />

J Craniofac Surg 8(1):17, 1997.<br />

431. Currarina G, Silverman FN: Orbital hypotelorism,<br />

arhinencephaly and trigonocephaly. Radiology 74:206,<br />

1960.<br />

432. Evereklioglu C, Doganay S, Hamdi ER, et al: Interpupillary<br />

index: a new parameter for hypo-hypertelorism. J<br />

Craniomaxillofac Surg 29:191, 2001.<br />

433. Converse JM et al: Ocular hypertelorism and<br />

pseudohypertelorism. Advances in surgical treatment.<br />

Plast Reconstr Surg 45:1, 1970.<br />

434. McCarthy JG et al: Hypertelorism correction in the young<br />

child. Plast Reconstr Surg 86:214, 1990.<br />

435. Ortiz Monasterio F, Medina O, Musolas A: Geometrical<br />

planning for the correction of orbital hypertelorism. Plast<br />

Reconstr Surg 86:650, 1990.<br />

436. Van der Meulen JC, Vaandrager J: <strong>Surgery</strong> related to the<br />

correction of hypertelorism. Plast Reconstr Surg 71:6,<br />

1983.<br />

437. Kennedy RE: The effect of early enucleation on the orbit.<br />

Am J Ophthalmol 73:80, 1965.<br />

438. Tessier P: Expansion chirurgicale de l’orbite. Les orbites<br />

trop petites. Exophthalmies Basedowiennes. Exorbitisme<br />

des dysostoses cranio-faciales. Atresies orbitaires des jeunes<br />

enuclees. Tumeurs orbitaires. Ann Chir Plast 14:207,<br />

1969.<br />

439. Marchac D et al: Orbital expansion for anophthalmia and<br />

microorbitism. Plast Reconstr Surg 59:486, 1977.<br />

440. Elisevich K, Bite U, Colcleugh R: Microorbitalism: A<br />

technique for orbital rim expansion. Plast Reconstr Surg<br />

88:609, 1991.<br />

441. Lo AKM et al: The role of tissue expanders in an anophthalmic<br />

animal model. Plast Reconstr Surg 86:399, 1990.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

442. Rodallec A et al: Anophthalmie congenitale. Controle de<br />

l’osteogenese par prothese expansive intraorbitaire. J Fr<br />

Ophthalmol 11:661, 1988.<br />

443. Drommer RB: The history of the “Le Fort I osteotomy”. J<br />

Maxillofac Surg 14:119, 1986.<br />

444. Munro IR, Beals SP, Griffin GJ: The self-retained Le Fort I<br />

osteotomy. Plast Reconstr Surg 80:843, 1987.<br />

445. Bell WH, Levy BM: Revascularization and bone healing<br />

following total maxillary osteotomy. J Dent Res 82:96,<br />

1973.<br />

446. Pepersack WJ: Tooth vitality after alveolar segmental<br />

osteotomy. J Maxillofac Surg 1:85, 1973.<br />

447. Henderson D, Jackson IT: Naso-maxillary hypoplasia—<br />

The Le Fort II osteotomy. Br J Oral Surg 2:77, 1973.<br />

448. Converse JM et al: The treatment of nasomaxillary hypoplasia.<br />

A new pyramidal naso-orbital maxillary osteotomy.<br />

Plast Reconstr Surg 45:527, 1970.<br />

449. Bachmayer DI, Ross RB, Munro IR: Maxillary growth<br />

following Le Fort III advancement surgery in Crouzon,<br />

Apert, and Pfeiffer syndromes. Am J Orthod Dentofac<br />

Orthop 90:420-430, 1986.<br />

450. Kaban LB et al: Midface position after Le Fort III advancement.<br />

Plast Reconstr Surg 73:758, 1984.<br />

451. McCarthy JG et al: Le Fort III advancement osteotomy in<br />

the growing child. Plast Reconstr Surg 74:343, 1984.<br />

452. McCarthy JG et al: The Le Fort III advancement osteotomy<br />

in the child under 7 years of age. Plast Reconstr Surg<br />

86:633, 1990.<br />

453. Whitaker LA et al: Combined report of problems and<br />

complications in 793 craniofacial operations. Plast Reconstr<br />

Surg 64:198, 1979.<br />

454. Munro IR, Sabatier RE: An analysis of 12 years of<br />

craniomaxillofacial surgery in Toronto. Plast Reconstr Surg<br />

76:29, 1985.<br />

455. David DJ, Cooter RD: <strong>Craniofacial</strong> infection in 10 years of<br />

transcranial surgery. Plast Reconstr Surg 80:213, 1987.<br />

456. Poole MD: Complications in craniofacial surgery. Br J Plast<br />

Surg 41:608, 1988.<br />

457. Codovilla A: On a means of lengthening in the lower limbs,<br />

the muscles and tissues that are shortened through deformity.<br />

Am J Orthop Surg 2:352, 1905.<br />

458. Ilizarov GA: The principles of the Ilizarov method. Bull<br />

Hosp Joint Dis Orthop Inst 48:1, 1988.<br />

459. Ilizarov GA: A new principle of osteosynthesis with the use<br />

of crossing pins and rings. In: Collection of scientific<br />

works of the Kurgan Regional Scientific Medical Society.<br />

Kurgan Med Soc 145-160, 1954.<br />

460. Snyder CC, Levine GA, Swanson HM, Browne EZ Jr:<br />

Mandibular lengthening by gradual distraction. Preliminary<br />

report. Plast Reconstr Surg 51:506, 1973.<br />

461. McCarthy JG, Stelnicki EJ, Grayson BH: Distraction osteogenesis<br />

of the mandible: a ten-year experience. Semin<br />

Orthod 5:3, 1999.<br />

462. Habal MB: A future domain distractor for the facial<br />

skeleton. J Craniofac Surg 4:414, 1995.<br />

463. Polley JW, Cohen M, Reisberg D, et al: Monobloc<br />

craniomaxillofacial distraction osteogenesis in a newborn<br />

with severe craniofacial synostosis: a preliminary report. J<br />

Craniofac Surg 6:421, 1995.<br />

464. Chin M, Toth BA: Le Fort III advancement with gradual<br />

distraction using internal devices. Plast Reconstr Surg<br />

100:819, 1997.<br />

61


SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

465. Tavakoli K, Walsh WR, Bonar F, et al: The role of latency<br />

in mandibular osteodistraction. J Craniomaxillofac Surg<br />

26:209, 1998.<br />

466. Cedars MG, Linck DL II, Chin M, Toth BA: Advancement<br />

of the midface using distraction techniques. Plast Reconstr<br />

Surg 103:429, 1999.<br />

467. Farhadieh RD, Gianoutsos MP, Dickinson R, Walsh WR:<br />

Effect of distraction rate on biomechanical, mineralization,<br />

and histologic properties of an ovine mandible model.<br />

Plast Reconstr Surg 105:889, 2000.<br />

468. Troulis MJ, Glowacki J, Perrott DH, Kaban LB: Effects of<br />

latency and rate on bone formation in a porcine mandibular<br />

distraction model. J Oral Maxillofac Surg 58:507, 2000.<br />

469. Ilizarov GA: Clinical application of the tension-stress effect<br />

for limb lengthening. Clin Orthop Rel Res 250:8, 1990.<br />

470. Aronson J: Experimental and clinical experience with distraction<br />

osteogenesis. Cleft Palate Craniofac J 31:473, 1994.<br />

471. Rachmiel A, Laufer D, Jackson IT, et al: Midface membranous<br />

bone lengthening: a one-year histological and morphological<br />

follow-up of distraction osteogenesis. Calcif<br />

Tissue Int 62:370, 1998.<br />

472. Smith SW, Sachdeva RCL, Cope JB: Evaluation of the<br />

consolidation period during osteodistraction using computed<br />

tomography. Am J Orthod Dentofacial Orthop<br />

116:254, 1999.<br />

473. Felemovicius J, Ortiz Monasterio F, Gomez Radillo LS,<br />

Serna A: Determining the optimal time for consolidation<br />

after distraction osteogenesis. J Craniofac Surg 11:430,<br />

2000.<br />

474. Molina F: Discussion of “<strong>Craniofacial</strong> distraction osteogenesis:<br />

a review of 3278 cases” by Mofid MM, Manson PN,<br />

Robertson BC, et al. Plast Reconstr Surg 108:1115, 2001.<br />

475. Tharanon W, Sinn DP: Mandibular distraction osteogenesis<br />

with multidirectional extraoral distraction device in<br />

hemifacial microsomia patients: three-dimensional treatment<br />

planning, prediction tracings, and case outcomes. J<br />

Craniofac Surg 10:202, 1999.<br />

476. Gateno J, Teichgraeber JF, Aguilar E: Computer planning<br />

for distraction osteogenesis. Plast Reconstr Surg 105:873,<br />

2000.<br />

477. Gateno J, Allen ME, Teichgraeber JF, Messersmith ML: An<br />

in vitro study of the accuracy of a new protocol for planning<br />

distraction osteogenesis of the mandible. J Oral Maxillofac<br />

Surg 58:985, 2000.<br />

478. Pensler JM, Goldberg DP, Lindell B, Carroll NC: Skeletal<br />

distraction of the hypoplastic mandible. Ann Plast Surg<br />

34:130, 1995.<br />

479. McCarthy JG, Williams JK, Grayson BH, Crombie JS: Controlled<br />

multiplanar distraction of the mandible: device<br />

development and clinical application. J Craniofac Surg<br />

9:322, 1998.<br />

480. Douglas LR, Douglass JB, Nakeeb S, et al: Intraoral distraction<br />

osteogenesis in the baboon mandible using a tooth and<br />

bone-anchored appliance. J Oral Maxillofac Surg 58:49,<br />

2000.<br />

481. Guerrero CA, Bell WH, Contasti GI, Rodriguez AM: Intraoral<br />

mandibular distraction osteogenesis. Semin Orthod<br />

5:35, 1999.<br />

482. Hollier LH, Gosain A, Stelnicki E, et al: <strong>Craniofacial</strong><br />

distraction osteogenesis. J Craniofac Surg 10:268, 1999.<br />

483. Rachmiel A, Aizenbud D, Eleftheriou S, et al: Extraoral vs.<br />

intraoral distraction osteogenesis in the treatment of hemifacial<br />

microsomia. Ann Plast Surg 45:386, 2000.<br />

62<br />

484. Polley JW, Figueroa AA, Chardel FT, et al: Monobloc<br />

craniomaxillofacial distraction osteogenesis in a newborn<br />

with severe craniofacial synostosis: a preliminary report. J<br />

Craniofac Surg 6:421, 1995.<br />

485. Cohen SR: <strong>Craniofacial</strong> distraction with a modular internal<br />

distraction system: evolution of design and surgical techniques.<br />

Plast Reconstr Surg 103:1592, 1999.<br />

486. Cohen SR, Holmes RE, Amis P, Fichtner H: <strong>Internal</strong><br />

craniofacial distraction with biodegradable devices: early<br />

stabilization and protected bone regeneration. J Craniofac<br />

Surg 11:354, 2000.<br />

487. Cohen SR, Holmes RE: <strong>Internal</strong> Le Fort III distraction with<br />

biodegradable devices. J Craniofac Surg 12:264, 2001.<br />

488. Schmelzeisen R, Neumann G, Fecht R von der: Distraction<br />

osteogenesis in the mandible with a motor-driven plate: a<br />

preliminary animal study. Br J Oral Maxillofac Surg 34:375,<br />

1996.<br />

489. Swennen G, Schliephake H, Dempf R, et al: <strong>Craniofacial</strong><br />

distraction osteogenesis: a review of the literature. <strong>Part</strong> I:<br />

clinical studies. Int J Oral Maxillofac Surg 30:89, 2001.<br />

490. Lauritzen C, Munro IR, Ross RB: Classification and treatment<br />

of hemifacial microsomia. Scand J Plast Reconstr<br />

Surg 19:33, 1985.<br />

491. Vento AR, LaBrie RA, Mulliken JB: The O.M.E.N.S. classification<br />

of hemifacial microsomia. Cleft Palate Craniofac<br />

J 28:68, 1991.<br />

492. Munro IR: One-stage reconstruction of the temporomandibular<br />

joint in hemifacial microsomia. Plast Reconstr Surg<br />

66:6999, 1980.<br />

493. McCarthy JG: The role of distraction osteogenesis in the<br />

reconstruction of the mandible in unilateral craniofacial<br />

microsomia. Clin Plast Surg 21:625, 1994.<br />

494. McCormick SU, McCarthy JG, Grayson BH, et al: Effect<br />

of mandibular distraction on the temporomandibular<br />

joint: <strong>Part</strong> 1, canine study. J Craniofac Surg 6(5):358,<br />

1995.<br />

495. McCormick SU, Grayson BH, McCarthy JG, Staffenberg D:<br />

Effect of mandibular distraction on the temporomandibular<br />

joint: <strong>Part</strong> 2, clinical study. J Craniofac Surg 6(5):364,<br />

1995.<br />

496. Arnett GW: A redefinition of bilateral sagittal osteotomy<br />

(BSO) advancement relapse. Am J Orthod Dentofac Orthop<br />

104:506, 1993.<br />

497. Ellis E III, Hinton RJ: Histologic examination of the temporomandibular<br />

joint after mandibular advancement with<br />

and without rigid fixation. J Oral Maxillofac Surg 49:1316,<br />

1991.<br />

498. McNeill C, ed: Temporomandibular Disorders: Guidelines<br />

for Classification, Assessment, and Management.<br />

Chicago, Quintessence Publ, 1993, p 27-38.<br />

499. Enlow DH: Handbook of Facial Growth. Philadelphia,<br />

WB Saunders, 1975, p 25-57.<br />

500. Moss ML, Salentijin L: The primary role functional matrices<br />

in facial growth. Am J Orthod 55:566, 1969.<br />

501. Guyette TW, Polley JW, Figueroa A, et al: Changes in<br />

speech following unilateral mandibular distraction osteogenesis<br />

in patients with hemifacial microsomia. Cleft<br />

Palate Craniofac J 38:179, 2001.<br />

502. Hoffmeister B, Marcks C, Wolff K: The floating bone<br />

concept in intraoral mandibular distraction. Presented at<br />

the 14 th Congress of the European Association for Cranio-<br />

Maxillofacial <strong>Surgery</strong>, Helsinki, Finland, 1998. J<br />

Craniomaxillofac Surg 26(Suppl 1):76, 1998.


503. Kunz C, Hammer B, Prein J: Manipulation of callus after<br />

linear distraction: a “lifeboat” or an alternative to<br />

multivectorial distraction osteogenesis of the mandible?<br />

Plast Reconstr Surg 105:674, 2000.<br />

504. Denny A, Kalantarian B: Mandibular distraction in neonates:<br />

a strategy to avoid tracheostomy. Plast Reconstr<br />

Surg 109:896, 2002.<br />

505. Ortiz Monasterio F, Molina F, Andrade L, et al: Simultaneous<br />

mandibular and maxillary distraction in hemifacial<br />

microsomia in adults: avoidal occlusal disasters. Plast<br />

Reconstr Surg 100:852, 1997.<br />

506. Molina F, Ortiz Monasterio F: Maxillary distraction: Three<br />

years of clinical experience. Presented at the Annual<br />

Meeting of the American Society of <strong>Plastic</strong> and Reconstructive<br />

Surgeons, Dallas, Texas, November 1996.<br />

507. Ko EW-C, Figueroa AA, Polley JW: Soft tissue profile<br />

changes after maxillary advancement with distraction osteogenesis<br />

by use of a rigid external distraction device: a<br />

1-year follow-up. J Oral Maxillofac Surg 58:959, 2000.<br />

508. Ko EW-C, Figueroa AA, Guyette TW, et al: Velopharyngeal<br />

changes after maxillary advancement in cleft patients with<br />

distraction osteogenesis using a rigid external distraction<br />

device: a 1-year cephalometric follow-up. J Craniofac<br />

Surg 10:312, 1999.<br />

509. Cohen SR, Rutrick RE, Burstein FD: Distraction osteogenesis<br />

of the human craniofacial skeleton: initial experience with<br />

a new distraction system. J Craniofac Surg 6(5):368, 1995.<br />

510. Meling TR, Tveten S, Due-Tonnessen BJ, et al: Monobloc<br />

and midface distraction osteogenesis in pediatric patients<br />

with severe syndromal craniosynostosis. Pediatr Neurosurg<br />

33:89, 2000.<br />

511. Gosain AK, Hanson P: Case reports in midfacial distraction:<br />

in search of the optimal technique. In: Chen YR (ed),<br />

<strong>Craniofacial</strong> <strong>Surgery</strong> VIII. Bologna, Monduzzi Editore,<br />

2002.<br />

512. Fearon JA: The Le Fort III osteotomy: to distract or not to<br />

distract? Plast Reconstr Surg 107:1091, 2001.<br />

513. Nadal E, Dogliotti PLV, Rodriguez JC, Zuccaro G: <strong>Craniofacial</strong><br />

distraction osteogenesis en bloc. J Craniofac Surg<br />

11:246, 2000.<br />

514. Cohen SR, Boydston W, Hudgins R, Burstein FD: Monobloc<br />

and facial bipartition distraction with internal devices. J<br />

Craniofac Surg 10:244, 1999.<br />

515. Hierl T, Hemprich A: Callus distraction of the midface in<br />

the severely atrophied maxilla—a case report. Cleft Palate<br />

Craniofac J 36:457, 1999.<br />

516. Komuro Y, Akizuki T, Kurakata M, Ohmori K: Histological<br />

examination of regenerated bone through craniofacial<br />

bone distraction in clinical studies. J Craniofac Surg 10:308,<br />

1999.<br />

517. Glat PM, Staffenberg DA, Karp NS, et al: Multidimensional<br />

distraction osteogenesis: the canine zygoma. Plast Reconstr<br />

Surg 94:753, 1994.<br />

518. Liou EJ, Chen PK, Huang CS, Chen YR: Interdental<br />

distraction osteogenesis and rapid orthodontic tooth movement:<br />

a novel approach to approximate a wide alveolar<br />

cleft or bony defect. Plast Reconstr Surg 105:1262, 2000.<br />

SRPS Volume 10, Number 17, <strong>Part</strong> 2<br />

519. Kobayashi S, Honda T, Saitch A, Kashiwa K: Unilateral<br />

coronal synostosis treated by internal forehead distraction.<br />

J Craniofac Surg 10:467, 1999.<br />

520. Matsumoto K, Nakanishi H, Seike T, et al: Application of<br />

the distraction technique to scaphocephaly. J Craniofac<br />

Surg 11:172, 2000.<br />

521. Mofid MM, Manson PN, Robertson BC, et al: <strong>Craniofacial</strong><br />

distraction osteogenesis: a review of 3278 cases. Plast<br />

Reconstr Surg 108:1103, 2001.<br />

522. Stein SC, Schut L: Management of scaphocephaly. Surg<br />

Neurol 7:153, 1977.<br />

523. Wolfe SA: Autogenous bone grafting of orbitocranial<br />

defects versus the use of alloplastic materials. In: Caronni<br />

EP (ed), <strong>Craniofacial</strong> <strong>Surgery</strong>. Boston, Little Brown,<br />

1982. Ch 41, pp 476- 489.<br />

524. Lipschitz AH, Kenkel JM: Implantation: Bone, Cartilage,<br />

and Alloplasts. Selected Read Plast Surg 10(2), 2004.<br />

525. Cutting CB, McCarthy JG, Berenstein A: Blood supply of<br />

the upper craniofacial skeleton: The search for composite<br />

calvarial bone flaps.. Plast Reconstr Surg 74:603,<br />

1984.<br />

526. Spear SL, Wiegering CE: Temporal fossa bone grafts: A<br />

new technique in craniofacial surgery. Plast Reconstr Surg<br />

79:531, 1987.<br />

527. Bite U et al: Vascularized skull bone grafts in craniofacial<br />

surgery. Ann Plast Surg 19:3, 1987.<br />

528. Antonyshin O, Colcleugh RG, Anderson C: Growth potential<br />

in suture bone inlay grafts: A comparison of vascularized<br />

and free calvarial bone grafts. Plast Reconstr Surg<br />

79:1, 1987.<br />

529. Byrd HS, Hobar PC, Shewmake K: Augmentation of the<br />

craniofacial skeleton with porous hydroxyapatite granules.<br />

Plast Reconstr Surg 91:15, 1993.<br />

530. Burstein FD, Cohen SR, Hudgins R, et al: The use of<br />

hydroxyapatite cement in secondary craniofacial reconstruction.<br />

Plast Reconstr Surg 104:1270, 1999.<br />

531. Jackson IT, Yavuzer R: Hydroxyapatite cement: an alternative<br />

for craniofacial skeletal contour refinements. Br J<br />

Plast Surg 53:24, 2000.<br />

532. Jackson IT, Carls F, Bush K, et al: Assessment and<br />

treatment of facial deformity resulting from radiation to<br />

the orbital area in childhood. Plast Reconstr Surg 98:1169,<br />

1996.<br />

533. Nwoku AL, Koch H: Effect of radiation injury on the<br />

growing face. J Maxillofac Surg 3:28, 1975.<br />

534. Larson EP: Long-term effects of radiation therapy in the<br />

head and neck. Clin Plast Surg 20:485, 1993.<br />

535. Marx RE, Johnson RP: Studies in the radiobiology of<br />

osteoradionecrosis and their clinical significance. Oral<br />

Surg 64:379, 1987.<br />

536. Guyuron B: The hourglass facial deformity. J Craniomaxillofac<br />

Surg 18:187, 1990.<br />

537. Guyuron B, Dagys AP, Munro IR: Long-term effects of<br />

orbital irradiation. Head neck Surg 10:85, 1987.<br />

538. Kawamoto HK Jr: Elective osteotomies and bone grafting<br />

of irradiated midfacial bones. J Craniomaxillofac Surg<br />

15:199, 1987.<br />

63

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!