A malignant neoplasm of the ADRENAL CORTEX. Adrenocortical carcinomas are unencapsulated anaplastic (ANAPLASIA) masses sometimes exceeding 20 cm or 200 g. They are more likely to be functional than nonfunctional, and produce ADRENAL CORTEX HORMONES that may result in hypercortisolism (CUSHING SYNDROME); HYPERALDOSTERONISM; and/or VIRILISM.
Tumors or cancers of the ADRENAL CORTEX.
A derivative of the insecticide DICHLORODIPHENYLDICHLOROETHANE that specifically inhibits cells of the adrenal cortex and their production of hormones. It is used to treat adrenocortical tumors and causes CNS damage, but no bone marrow depression.
A benign neoplasm of the ADRENAL CORTEX. It is characterized by a well-defined nodular lesion, usually less than 2.5 cm. Most adrenocortical adenomas are nonfunctional. The functional ones are yellow and contain LIPIDS. Depending on the cell type or cortical zone involved, they may produce ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and/or ANDROSTENEDIONE.
The outer layer of the adrenal gland. It is derived from MESODERM and comprised of three zones (outer ZONA GLOMERULOSA, middle ZONA FASCICULATA, and inner ZONA RETICULARIS) with each producing various steroids preferentially, such as ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and ANDROSTENEDIONE. Adrenal cortex function is regulated by pituitary ADRENOCORTICOTROPIN.
A malignant neoplasm made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases. It is a histological type of neoplasm but is often wrongly used as a synonym for "cancer." (From Dorland, 27th ed)
A condition caused by prolonged exposure to excess levels of cortisol (HYDROCORTISONE) or other GLUCOCORTICOIDS from endogenous or exogenous sources. It is characterized by upper body OBESITY; OSTEOPOROSIS; HYPERTENSION; DIABETES MELLITUS; HIRSUTISM; AMENORRHEA; and excess body fluid. Endogenous Cushing syndrome or spontaneous hypercortisolism is divided into two groups, those due to an excess of ADRENOCORTICOTROPIN and those that are ACTH-independent.
Tumors or cancer of the ADRENAL GLANDS.
An anterior pituitary hormone that stimulates the ADRENAL CORTEX and its production of CORTICOSTEROIDS. ACTH is a 39-amino acid polypeptide of which the N-terminal 24-amino acid segment is identical in all species and contains the adrenocorticotrophic activity. Upon further tissue-specific processing, ACTH can yield ALPHA-MSH and corticotrophin-like intermediate lobe peptide (CLIP).
The main glucocorticoid secreted by the ADRENAL CORTEX. Its synthetic counterpart is used, either as an injection or topically, in the treatment of inflammation, allergy, collagen diseases, asthma, adrenocortical deficiency, shock, and some neoplastic conditions.
A rare benign tumor of the adrenal gland, several centimeters in diameter, composed in varying proportions of adipose tissue, lymphocytes, and primitive myeloid cells, probably a developmental abnormality. (Dorland, 27th ed)
A pair of glands located at the cranial pole of each of the two KIDNEYS. Each adrenal gland is composed of two distinct endocrine tissues with separate embryonic origins, the ADRENAL CORTEX producing STEROIDS and the ADRENAL MEDULLA producing NEUROTRANSMITTERS.
Neoplasm derived from displaced cells (rest cells) of the primordial ADRENAL GLANDS, generally in patients with CONGENITAL ADRENAL HYPERPLASIA. Adrenal rest tumors have been identified in TESTES; LIVER; and other tissues. They are dependent on ADRENOCORTICOTROPIN for growth and adrenal steroid secretion.
Excision of one or both adrenal glands. (From Dorland, 28th ed)
A microsomal cytochrome P450 enzyme that catalyzes the 17-alpha-hydroxylation of progesterone or pregnenolone and subsequent cleavage of the residual two carbons at C17 in the presence of molecular oxygen and NADPH-FERRIHEMOPROTEIN REDUCTASE. This enzyme, encoded by CYP17 gene, generates precursors for glucocorticoid, androgen, and estrogen synthesis. Defects in CYP17 gene cause congenital adrenal hyperplasia (ADRENAL HYPERPLASIA, CONGENITAL) and abnormal sexual differentiation.
A carcinoma derived from stratified SQUAMOUS EPITHELIAL CELLS. It may also occur in sites where glandular or columnar epithelium is normally present. (From Stedman, 25th ed)
Pathological processes of the ADRENAL CORTEX.
A primary malignant neoplasm of epithelial liver cells. It ranges from a well-differentiated tumor with EPITHELIAL CELLS indistinguishable from normal HEPATOCYTES to a poorly differentiated neoplasm. The cells may be uniform or markedly pleomorphic, or form GIANT CELLS. Several classification schemes have been suggested.
A benign epithelial tumor with a glandular organization.
Development of female secondary SEX CHARACTERISTICS in the MALE. It is due to the effects of estrogenic metabolites of precursors from endogenous or exogenous sources, such as ADRENAL GLANDS or therapeutic drugs.
Excess production of ADRENAL CORTEX HORMONES such as ALDOSTERONE; HYDROCORTISONE; DEHYDROEPIANDROSTERONE; and/or ANDROSTENEDIONE. Hyperadrenal syndromes include CUSHING SYNDROME; HYPERALDOSTERONISM; and VIRILISM.
Steroids that contain a ketone group at position 17.
A metabolite of PROGESTERONE with a hydroxyl group at the 17-alpha position. It serves as an intermediate in the biosynthesis of HYDROCORTISONE and GONADAL STEROID HORMONES.
Rare autosomal dominant syndrome characterized by mesenchymal and epithelial neoplasms at multiple sites. MUTATION of the p53 tumor suppressor gene, a component of the DNA DAMAGE response pathway, apparently predisposes family members who inherit it to develop certain cancers. The spectrum of cancers in the syndrome was shown to include, in addition to BREAST CANCER and soft tissue sarcomas (SARCOMA); BRAIN TUMORS; OSTEOSARCOMA; LEUKEMIA; and ADRENOCORTICAL CARCINOMA.
Surgery performed on any endocrine gland.
Histochemical localization of immunoreactive substances using labeled antibodies as reagents.
A hormone secreted by the ADRENAL CORTEX that regulates electrolyte and water balance by increasing the renal retention of sodium and the excretion of potassium.
A transcription factor and member of the nuclear receptor family NR5 that is expressed throughout the adrenal and reproductive axes during development. It plays an important role in sexual differentiation, formation of primary steroidogenic tissues, and their functions in post-natal and adult life. It regulates the expression of key steroidogenic enzymes.
Catalyze the oxidation of 3-hydroxysteroids to 3-ketosteroids.
Adrenal cortex hormones are steroid hormones produced by the outer portion of the adrenal gland, consisting of glucocorticoids, mineralocorticoids, and androgens, which play crucial roles in various physiological processes such as metabolism regulation, stress response, electrolyte balance, and sexual development and function.
Molecular products metabolized and secreted by neoplastic tissue and characterized biochemically in cells or body fluids. They are indicators of tumor stage and grade as well as useful for monitoring responses to treatment and predicting recurrence. Many chemical groups are represented including hormones, antigens, amino and nucleic acids, enzymes, polyamines, and specific cell membrane proteins and lipids.
An enzyme that catalyzes the desaturation (aromatization) of the ring A of C19 androgens and converts them to C18 estrogens. In this process, the 19-methyl is removed. This enzyme is membrane-bound, located in the endoplasmic reticulum of estrogen-producing cells of ovaries, placenta, testes, adipose, and brain tissues. Aromatase is encoded by the CYP19 gene, and functions in complex with NADPH-FERRIHEMOPROTEIN REDUCTASE in the cytochrome P-450 system.
A cell line derived from cultured tumor cells.
A lesion with cytological characteristics associated with invasive carcinoma but the tumor cells are confined to the epithelium of origin, without invasion of the basement membrane.
A major C19 steroid produced by the ADRENAL CORTEX. It is also produced in small quantities in the TESTIS and the OVARY. Dehydroepiandrosterone (DHEA) can be converted to TESTOSTERONE; ANDROSTENEDIONE; ESTRADIOL; and ESTRONE. Most of DHEA is sulfated (DEHYDROEPIANDROSTERONE SULFATE) before secretion.
A group of polycyclic compounds closely related biochemically to TERPENES. They include cholesterol, numerous hormones, precursors of certain vitamins, bile acids, alcohols (STEROLS), and certain natural drugs and poisons. Steroids have a common nucleus, a fused, reduced 17-carbon atom ring system, cyclopentanoperhydrophenanthrene. Most steroids also have two methyl groups and an aliphatic side-chain attached to the nucleus. (From Hawley's Condensed Chemical Dictionary, 11th ed)
Tomography using x-ray transmission and a computer algorithm to reconstruct the image.
A prediction of the probable outcome of a disease based on a individual's condition and the usual course of the disease as seen in similar situations.
Unanticipated information discovered in the course of testing or medical care. Used in discussions of information that may have social or psychological consequences, such as when it is learned that a child's biological father is someone other than the putative father, or that a person tested for one disease or disorder has, or is at risk for, something else.
A malignant neoplasm characterized by the formation of numerous, irregular, finger-like projections of fibrous stroma that is covered with a surface layer of neoplastic epithelial cells. (Stedman, 25th ed)
Cells grown in vitro from neoplastic tissue. If they can be established as a TUMOR CELL LINE, they can be propagated in cell culture indefinitely.
Antineoplastic agents that are used to treat hormone-sensitive tumors. Hormone-sensitive tumors may be hormone-dependent, hormone-responsive, or both. A hormone-dependent tumor regresses on removal of the hormonal stimulus, by surgery or pharmacological block. Hormone-responsive tumors may regress when pharmacologic amounts of hormones are administered regardless of whether previous signs of hormone sensitivity were observed. The major hormone-responsive cancers include carcinomas of the breast, prostate, and endometrium; lymphomas; and certain leukemias. (From AMA Drug Evaluations Annual 1994, p2079)
Methods which attempt to express in replicable terms the extent of the neoplasm in the patient.
RNA sequences that serve as templates for protein synthesis. Bacterial mRNAs are generally primary transcripts in that they do not require post-transcriptional processing. Eukaryotic mRNA is synthesized in the nucleus and must be exported to the cytoplasm for translation. Most eukaryotic mRNAs have a sequence of polyadenylic acid at the 3' end, referred to as the poly(A) tail. The function of this tail is not known for certain, but it may play a role in the export of mature mRNA from the nucleus as well as in helping stabilize some mRNA molecules by retarding their degradation in the cytoplasm.
Retroperitoneal neoplasms are a diverse group of tumors that originate in the retroperitoneal space, which is the area behind the peritoneum and includes the kidneys, adrenal glands, pancreas, and major blood vessels.
Glycoproteins that inhibit pituitary FOLLICLE STIMULATING HORMONE secretion. Inhibins are secreted by the Sertoli cells of the testes, the granulosa cells of the ovarian follicles, the placenta, and other tissues. Inhibins and ACTIVINS are modulators of FOLLICLE STIMULATING HORMONE secretions; both groups belong to the TGF-beta superfamily, as the TRANSFORMING GROWTH FACTOR BETA. Inhibins consist of a disulfide-linked heterodimer with a unique alpha linked to either a beta A or a beta B subunit to form inhibin A or inhibin B, respectively
Any of the processes by which nuclear, cytoplasmic, or intercellular factors influence the differential control of gene action in neoplastic tissue.
Evaluation undertaken to assess the results or consequences of management and procedures used in combating disease in order to determine the efficacy, effectiveness, safety, and practicability of these interventions in individual cases or series.
Death resulting from the presence of a disease in an individual, as shown by a single case report or a limited number of patients. This should be differentiated from DEATH, the physiological cessation of life and from MORTALITY, an epidemiological or statistical concept.

YM116, 2-(1H-imidazol-4-ylmethyl)-9H-carbazole, decreases adrenal androgen synthesis by inhibiting C17-20 lyase activity in NCI-H295 human adrenocortical carcinoma cells. (1/185)

The concentrations of androstenedione and dehydroepiandrosterone, products of C17-20 lyase, in the medium after a 6-hr incubation of NCI-H295 cells were decreased by YM116 (2-(1H-imidazol-4-ylmethyl)-9H-carbazole) (IC50: 3.6 and 2.1 nM) and ketoconazole (IC50: 54.9 and 54.2 nM). 17Alpha-hydroxyprogesterone, a product of 17alpha-hydroxylase, was increased by YM116 (1-30 nM) and by ketoconazole (10-300 nM) and then was decreased at higher concentrations of both agents (IC50: 180 nM for YM116, 906 nM for ketoconazole), indicating that YM116 and ketoconazole were 50- and 16.5-fold more specific inhibitors of C17-20 lyase, respectively, than 17alpha-hydroxylase. Compatible with these findings, progesterone, a substrate of 17alpha-hydroxylase, was increased by these agents. Cortisol production was inhibited by YM116 and ketoconazole (IC50: 50.4 and 80.9 nM, respectively). YM116 was a 14-fold more potent inhibitor of androstenedione production than cortisol production, whereas ketoconazole was a nonselective inhibitor of the production of both steroids. YM116 and ketoconazole inhibited the C17-20 lyase activity in human testicular microsomes (IC50: 4.2 and 17 nM, respectively). These results demonstrate that YM116 reduces the synthesis of adrenal androgens by preferentially inhibiting C17-20 lyase activity.  (+info)

Comparison of expression and regulation of the high-density lipoprotein receptor SR-BI and the low-density lipoprotein receptor in human adrenocortical carcinoma NCI-H295 cells. (2/185)

In rodents, cholesterol for adrenal steroidogenesis is derived mainly from high-density lipoproteins (HDL) via the HDL receptor, scavenger receptor-BI (SR-BI). In humans cholesterol for steroidogenesis is considered to be derived from the low-density lipoprotein (LDL) receptor pathway, and the contribution of SR-BI to that is unknown. In the present study SR-BI expression and regulation by steroidogenic stimuli was analysed in human adrenocortical cells and compared with LDL receptor expression. In addition, the functional contribution of both receptors for cholesteryl ester delivery to human adrenocortical cells was compared. Northern blot and reverse transcription-PCR amplification and sequence analysis demonstrated the presence of SR-BI mRNA in foetal and adult human adrenal cortex. Furthermore, SR-BI mRNA was expressed to similar levels in human primary adrenocortical and adrenocortical carcinoma NCI-H295 cells, indicating its presence in the steroid-producing cells. Treatment of NCI-H295 cells with 8Br-cAMP, a stimulator of glucocorticoid synthesis via the protein kinase A second messenger signal transduction pathway, resulted in an increase of both SR-BI and LDL receptor mRNA levels in a time- and dose-dependent manner. The induction of SR-BI and LDL receptor by cAMP was independent of ongoing protein synthesis and occurred at the transcriptional level. Ligand blot experiments indicated that a protein of similar size to SR-BI is the major HDL-binding protein in NCI-H295 cells. Western blot analysis demonstrated that cAMP treatment increased the levels of LDL receptor and, to a lesser extent, SR-BI protein in NCI-H295 cells. Binding and uptake of cholesterol was quantitatively smaller from HDL than from LDL, both in basal as well as in cAMP-stimulated cells. Scatchard analysis under basal conditions indicated that NCI-H295 cells express twice as many specific binding sites for LDL than for HDL. Dissociation constant values (Kd; in nm) were approximately five times higher for HDL than for LDL, indicating a lower affinity of HDL compared with LDL. The combined effects of these two parameters and the low cholesteryl ester content of HDL subfraction 3 (HDL3) contributes to a lower cholesteryl ester uptake from HDL than from LDL by the NCI-H295 cells. In conclusion, both the SR-BI and LDL receptor genes are expressed in the human adrenal cortex and coordinately regulated by activators of glucocorticoid synthesis. In contrast to rodents, in human adrenocortical cells the HDL pathway of cholesterol delivery appears to be of lesser importance than the LDL pathway. Nevertheless, the SR-BI pathway may become of major importance in conditions of functional defects in the LDL receptor pathway.  (+info)

The expression of inhibin/activin subunits in the human adrenal cortex and its tumours. (3/185)

Inhibins and activins are dimeric proteins of the transforming growth factor-beta superfamily which have been shown to be expressed in the adrenal cortex. Recent studies have suggested a role for these peptides in the pathogenesis and/or function of adrenal tumours. To investigate further their physiological and pathological roles, we have documented immunoreactivity for inhibin alpha, betaA and betaB subunits in normal adult and fetal human adrenals, in hyperplastic adrenals and in adrenal tumours. In the normal and hyperplastic adult gland, diffuse immunopositivity was demonstrated for beta subunits, suggesting that activins (beta beta dimers) can be expressed in all zones. Inhibin alpha was limited to the zona reticularis and the innermost zona fasciculata in the normal gland, extending centripetally into the zona fasciculata in hyperplasia, supporting a role for ACTH in the regulation of expression, and suggesting that expression of inhibins (alpha beta dimers) is restricted. Immunopositivity for all three subunits was seen in both fetal and definitive zones of the fetal cortex, indicating that both inhibins and activins could be expressed in both. Immunopositivity for all three subunits was seen in most adrenocortical tumours. Loss of immunopositivity for inhibin alpha in a subgroup of carcinomas might indicate a role in tumour progression. The greater intensity of staining for inhibin alpha in tumours associated with Cushing's syndrome again suggests a link with cortisol production.  (+info)

Autocrine role of IGF-II in proliferation of human adrenocortical carcinoma NCI H295R cell line. (4/185)

In adrenocortical tumors, the malignant phenotype is associated with rearrangements (paternal isodisomy) at the 11p15 locus and IGF-II gene overexpression, strongly suggesting that the IGF system is a major determinant of adrenocortical tumor progression. The aim of this study was to validate an in vitro model for investigating the involvement of the IGF system in adrenocortical tumorigenesis. We analyzed the production of IGF mRNA and proteins, IGF-binding proteins (IGFBPs) and IGF receptors by the NCI H295R cell line, which is derived from a human adult adrenocortical carcinoma. H295R cells were shown to proliferate for a long period (26 days) in the absence of serum or any added growth factor. Northern blot analyses showed high IGF-II mRNA contents in H295R cells. The cells secreted large amounts of IGF-II protein (14 ng/10(6) cells per 48 h) although no IGF-I protein was detected. Western ligand blot analyses of conditioned media detected the presence of large amounts of a 34 kDa protein, which was identified as IGFBP-2 by immunoblotting. The presence of high-affinity binding sites for IGF-I and IGF-II on H295R cells was shown by binding experiments using radiolabeled IGFs and confirmed by reverse transcription PCR analyses showing type 1 and type 2 IGF receptors. Proliferation of H295R cells was inhibited by anti-IGF-II antibody (45%) and by anti-type 1 IGF receptor antibody (53%) indicating that IGF-II is an autocrine growth factor for these cells and that its effects are, at least in part, mediated by the type 1 IGF receptor. These findings confirm the involvement of the IGF system in adrenocortical tumors and suggest that the H295R cell line is a suitable in vitro model for studying the molecular mechanisms of adrenocortical tumor proliferation.  (+info)

Lack of response to octreotide in Cushing's syndrome due to metastatic adrenocortical carcinoma. (5/185)

Functional metastatic adrenocortical carcinoma is an uncommon cause of Cushing's syndrome, which rarely responds to conventional treatment. A patient presenting with Cushing's syndrome secondary to adrenocortical carcinoma underwent surgical resection. Postoperatively, she developed metastatic disease resistant to conventional chemotherapy. Octreotide, a somatostatin analogue which is effective in the treatment of several types of neuroendocrine tumour, was tried to ameliorate her secretory symptoms, but without any therapeutic effect.  (+info)

Analysis of genomic alterations in sporadic adrenocortical lesions. Gain of chromosome 17 is an early event in adrenocortical tumorigenesis. (6/185)

Genetic changes underlying the tumorigenesis of sporadic adrenocortical tumors are poorly characterized. To search for characteristic genomic imbalances involved in adrenocortical tumors, we examined 41 adrenocortical lesions (12 carcinomas, 23 adenomas, and 6 hyperplasias) by comparative genomic hybridization. Our results show that genetic alterations are more frequent in malignant than in benign lesions and that they rarely occur in hyperplastic lesions. The most frequent DNA copy number changes in adrenocortical carcinomas included losses of 1p21-31, 2q, 3p, 3q, 6q, 9p, and 11q14-qter, as well as gains and amplifications of 5q12, 9q32-qter, 12q, and 20q. The genomic aberrations prevalently occurring in adrenocortical adenomas were gains of 17q, 17p, and 9q32-qter. Gains found in 2 of 6 adrenocortical hyperplastic lesions involved chromosome 17 or 17q only. These data indicate that oncogenes determining the early tumorigenesis of adrenocortical tumors may exist on chromosome 17 and that the number of genomic alterations is closely associated with tumor behavior in adrenocortical tumors.  (+info)

Changes in neoplastic cell features and sensitivity to mitotane during mitotane-induced remission in a patient with recurrent, metastatic adrenocortical carcinoma. (7/185)

A 58-year-old man had adrenocortical carcinoma in the right adrenal gland. The tumour secreted excessive cortisol and dehydroepiandrosterone-sulphate (DHEA-S), and had invaded the right hepatic lobe and vena cava. Eleven months after surgical tumour resection, the serum DHEA-S levels again increased. Local tumour recurrence and a metastasis was found in the lung. Eleven months after surgery chemotherapy with mitotane (o,p'-DDD) was initiated. Twelve weeks of mitotane reduced serum DHEA-S levels and caused these tumours to disappear. The patient was then treated with low-dose mitotane (1.5-2.0 g/day) for 2 years. Serum levels of mitotane remained at less than 10 microg/ml. Although such low serum levels of mitotane and delayed initiation of mitotane after surgery have been proposed to weaken the antineoplastic effect of mitotane, the patient had a remission for 2 years. However, there was then local re-recurrence with an increase in serum DHEA-S and death 4 months later. The histological features of neoplastic cells were quite different comparing tumour resected at surgery and tumour at autopsy. The latter had more frequent mitotic nuclei. This tumour was initially sensitive to mitotane, but later became insensitive.  (+info)

Expression of inhibin alpha in adrenocortical tumours reflects the hormonal status of the neoplasm. (8/185)

Inhibins are gonadal glycoprotein hormones whose main endocrine function is to inhibit pituitary FSH secretion. In addition to testes and ovaries, other steroid-producing organs are sites of inhibin alpha subunit expression. To study the role of inhibins in human adrenal gland, we screened a panel of 150 adrenals (10 normal adrenals, 25 adrenocortical hyperplasias, 65 adrenocortical adenomas, 30 adrenocortical carcinomas and 20 phaeochromocytomas) for inhibin alpha expression. mRNA levels of inhibin alpha subunit were studied in 57 samples and all tissues were stained immunohistochemically with an inhibin alpha subunit-specific antibody. Inhibin alpha mRNA was detected in all adrenocortical tissues. Virilizing adenomas possessed a 10-fold higher median inhibin alpha mRNA expression than did normal adrenals. Bilaterally and nodularly hyperplastic adrenals and other than virilizing adrenocortical tumours had their median inhibin alpha mRNA levels close to those of normal adrenals. Immunohistochemically, inhibin alpha subunit was detectable in all normal and hyperplastic adrenals, as well as in 73% of the adrenocortical tumours. However, the percentage of inhibin alpha-positive cells varied greatly in different tumour types. The median percentage of positive cells was 10 in non-functional and Conn's adenomas, 30 in Cushing's adenomas and 75 in virilizing adenomas. In malignant adrenocortical tumours the median percentage of inhibin alpha-immunopositive cells was 20 in non-functional carcinomas, 30 in Conn's carcinomas, 65 in Cushing's carcinomas and 75 in virilizing carcinomas. All phaeochromocytomas were negative for inhibin alpha subunit both at the mRNA level and immunohistochemically. Our data show that inhibin alpha subunit is highly expressed in both normal and neoplastic androgen-producing adrenocortical cells, with less expression in cortisol-producing and hardly any in aldosterone-producing cells. This suggests a specific role for inhibins in the regulation of adrenal androgen production. We did not find any significant difference in inhibin alpha expression between benign and malignant adrenocortical tumours. Thus inhibin alpha gene does not seem to have a tumour suppressor role in human adrenal cortex.  (+info)

Adrenocortical carcinoma (ACC) is a rare cancer that develops in the outer layer of the adrenal gland, known as the adrenal cortex. The adrenal glands are small hormone-producing glands located on top of each kidney. They produce important hormones such as cortisol, aldosterone, and sex steroids.

ACC is a malignant tumor that can invade surrounding tissues and organs and may metastasize (spread) to distant parts of the body. Symptoms of ACC depend on the size and location of the tumor and whether it produces excess hormones. Common symptoms include abdominal pain, a mass in the abdomen, weight loss, and weakness. Excessive production of hormones can lead to additional symptoms such as high blood pressure, Cushing's syndrome, virilization (excessive masculinization), or feminization.

The exact cause of ACC is not known, but genetic factors, exposure to certain chemicals, and radiation therapy may increase the risk of developing this cancer. Treatment options for ACC include surgery, chemotherapy, radiation therapy, and targeted therapy. The prognosis for ACC varies depending on the stage and extent of the disease at diagnosis, as well as the patient's overall health.

Adrenal cortex neoplasms refer to abnormal growths (tumors) in the adrenal gland's outer layer, known as the adrenal cortex. These neoplasms can be benign or malignant (cancerous). Benign tumors are called adrenal adenomas, while cancerous tumors are called adrenocortical carcinomas.

Adrenal cortex neoplasms can produce various hormones, leading to different clinical presentations. For instance, they may cause Cushing's syndrome (characterized by excessive cortisol production), Conn's syndrome (caused by aldosterone excess), or virilization (due to androgen excess). Some tumors may not produce any hormones and are discovered incidentally during imaging studies for unrelated conditions.

The diagnosis of adrenal cortex neoplasms typically involves a combination of imaging techniques, such as CT or MRI scans, and hormonal assessments to determine if the tumor is functional or non-functional. In some cases, a biopsy may be necessary to confirm the diagnosis and differentiate between benign and malignant tumors. Treatment options depend on the type, size, location, and hormonal activity of the neoplasm and may include surgical excision, radiation therapy, chemotherapy, or a combination of these approaches.

Mitotane is not a general medical term, but it is a specific medication used in the treatment of certain types of cancer. Medically, Mitotane is known as an adrenal steroidogenesis inhibitor. It works by blocking the production of certain hormones in the adrenal glands, which can help to control the growth of some types of cancer cells.

Mitotane is primarily used in the treatment of advanced adrenocortical carcinoma (ACC), a rare type of cancer that develops in the outer layer of the adrenal gland. It may also be used in the management of other hormone-related cancers, such as certain types of thyroid or prostate cancer.

The medication is available in capsule form and is typically taken several times a day. Mitotane has a narrow therapeutic index, meaning that there is only a small range between an effective dose and a toxic one. Therefore, patients taking Mitotane require regular monitoring of their blood levels to ensure they remain within the safe range.

Common side effects associated with Mitotane include nausea, vomiting, diarrhea, dizziness, drowsiness, and changes in appetite or weight. More severe side effects may include neurological problems, such as confusion, disorientation, or memory loss, as well as an increased risk of infection due to its effect on the immune system.

An adrenocortical adenoma is a benign tumor that arises from the cells of the adrenal cortex, which is the outer layer of the adrenal gland. These tumors can produce and release various hormones, such as cortisol, aldosterone, or androgens, depending on the type of cells they originate from.

Most adrenocortical adenomas are nonfunctioning, meaning that they do not secrete excess hormones and may not cause any symptoms. However, some functioning adenomas can produce excessive amounts of hormones, leading to a variety of clinical manifestations. For example:

* Cortisol-secreting adenomas can result in Cushing's syndrome, characterized by weight gain, muscle wasting, thin skin, easy bruising, and mood changes.
* Aldosterone-producing adenomas can cause Conn's syndrome, marked by hypertension (high blood pressure), hypokalemia (low potassium levels), and metabolic alkalosis.
* Androgen-secreting adenomas may lead to hirsutism (excessive hair growth) or virilization (development of male secondary sexual characteristics) in women.

The diagnosis of an adrenocortical adenoma typically involves imaging tests, such as CT or MRI scans, and hormonal evaluations to determine if the tumor is functioning or not. Treatment usually consists of surgical removal of the tumor, especially if it is causing hormonal imbalances or growing in size.

The adrenal cortex is the outer portion of the adrenal gland, which is located on top of the kidneys. It plays a crucial role in producing hormones that are essential for various bodily functions. The adrenal cortex is divided into three zones:

1. Zona glomerulosa: This outermost zone produces mineralocorticoids, primarily aldosterone. Aldosterone helps regulate sodium and potassium balance and thus influences blood pressure by controlling the amount of fluid in the body.
2. Zona fasciculata: The middle layer is responsible for producing glucocorticoids, with cortisol being the most important one. Cortisol regulates metabolism, helps manage stress responses, and has anti-inflammatory properties. It also plays a role in blood sugar regulation and maintaining the body's response to injury and illness.
3. Zona reticularis: The innermost zone produces androgens, primarily dehydroepiandrosterone (DHEA) and its sulfate form (DHEAS). These androgens are weak compared to those produced by the gonads (ovaries or testes), but they can be converted into more potent androgens or estrogens in peripheral tissues.

Disorders related to the adrenal cortex can lead to hormonal imbalances, affecting various bodily functions. Examples include Addison's disease (insufficient adrenal cortical hormone production) and Cushing's syndrome (excessive glucocorticoid levels).

Carcinoma is a type of cancer that develops from epithelial cells, which are the cells that line the inner and outer surfaces of the body. These cells cover organs, glands, and other structures within the body. Carcinomas can occur in various parts of the body, including the skin, lungs, breasts, prostate, colon, and pancreas. They are often characterized by the uncontrolled growth and division of abnormal cells that can invade surrounding tissues and spread to other parts of the body through a process called metastasis. Carcinomas can be further classified based on their appearance under a microscope, such as adenocarcinoma, squamous cell carcinoma, and basal cell carcinoma.

Cushing syndrome is a hormonal disorder that occurs when your body is exposed to high levels of the hormone cortisol for a long time. This can happen due to various reasons such as taking high doses of corticosteroid medications or tumors that produce cortisol or adrenocorticotropic hormone (ACTH).

The symptoms of Cushing syndrome may include:

* Obesity, particularly around the trunk and upper body
* Thinning of the skin, easy bruising, and purple or red stretch marks on the abdomen, thighs, breasts, and arms
* Weakened bones, leading to fractures
* High blood pressure
* High blood sugar
* Mental changes such as depression, anxiety, and irritability
* Increased fatigue and weakness
* Menstrual irregularities in women
* Decreased fertility in men

Cushing syndrome can be diagnosed through various tests, including urine and blood tests to measure cortisol levels, saliva tests, and imaging tests to locate any tumors. Treatment depends on the cause of the condition but may include surgery, radiation therapy, chemotherapy, or adjusting medication dosages.

Adrenal gland neoplasms refer to abnormal growths or tumors in the adrenal glands. These glands are located on top of each kidney and are responsible for producing hormones that regulate various bodily functions such as metabolism, blood pressure, and stress response. Adrenal gland neoplasms can be benign (non-cancerous) or malignant (cancerous).

Benign adrenal tumors are called adenomas and are usually small and asymptomatic. However, some adenomas may produce excessive amounts of hormones, leading to symptoms such as high blood pressure, weight gain, and mood changes.

Malignant adrenal tumors are called adrenocortical carcinomas and are rare but aggressive cancers that can spread to other parts of the body. Symptoms of adrenocortical carcinoma may include abdominal pain, weight loss, and hormonal imbalances.

It is important to diagnose and treat adrenal gland neoplasms early to prevent complications and improve outcomes. Diagnostic tests may include imaging studies such as CT scans or MRIs, as well as hormone level testing and biopsy. Treatment options may include surgery, radiation therapy, chemotherapy, or a combination of these approaches.

Adrenocorticotropic Hormone (ACTH) is a hormone produced and released by the anterior pituitary gland, a small endocrine gland located at the base of the brain. ACTH plays a crucial role in the regulation of the body's stress response and has significant effects on various physiological processes.

The primary function of ACTH is to stimulate the adrenal glands, which are triangular-shaped glands situated on top of the kidneys. The adrenal glands consist of two parts: the outer cortex and the inner medulla. ACTH specifically targets the adrenal cortex, where it binds to specific receptors and initiates a series of biochemical reactions leading to the production and release of steroid hormones, primarily cortisol (a glucocorticoid) and aldosterone (a mineralocorticoid).

Cortisol is involved in various metabolic processes, such as regulating blood sugar levels, modulating the immune response, and helping the body respond to stress. Aldosterone plays a vital role in maintaining electrolyte and fluid balance by promoting sodium reabsorption and potassium excretion in the kidneys.

ACTH release is controlled by the hypothalamus, another part of the brain, which produces corticotropin-releasing hormone (CRH). CRH stimulates the anterior pituitary gland to secrete ACTH, which in turn triggers cortisol production in the adrenal glands. This complex feedback system helps maintain homeostasis and ensures that appropriate amounts of cortisol are released in response to various physiological and psychological stressors.

Disorders related to ACTH can lead to hormonal imbalances, resulting in conditions such as Cushing's syndrome (excessive cortisol production) or Addison's disease (insufficient cortisol production). Proper diagnosis and management of these disorders typically involve assessing the function of the hypothalamic-pituitary-adrenal axis and addressing any underlying issues affecting ACTH secretion.

Hydrocortisone is a synthetic glucocorticoid, which is a class of steroid hormones. It is identical to the naturally occurring cortisol, a hormone produced by the adrenal gland that helps regulate metabolism and helps your body respond to stress. Hydrocortisone has anti-inflammatory effects and is used to treat various inflammatory conditions such as allergies, skin disorders, and autoimmune diseases. It works by suppressing the immune system's response to reduce swelling, redness, itching, and other symptoms caused by inflammation.

Hydrocortisone is available in different forms, including oral tablets, topical creams, lotions, gels, and ointments, as well as injectable solutions. The specific use and dosage depend on the condition being treated and the individual patient's medical history and current health status.

As with any medication, hydrocortisone can have side effects, especially when used in high doses or for extended periods. Common side effects include increased appetite, weight gain, mood changes, insomnia, and skin thinning. Long-term use of hydrocortisone may also increase the risk of developing osteoporosis, diabetes, cataracts, and other health problems. Therefore, it is essential to follow your healthcare provider's instructions carefully when using this medication.

Myelolipoma is a type of benign tumor that occurs in the adrenal gland, which is located on top of each kidney. This tumor is composed of both fatty tissue (lipoma) and cells that are similar to those found in the bone marrow (myeloid). Myelolipomas are usually small and asymptomatic, but they can grow larger and cause symptoms such as abdominal pain or discomfort, depending on their size and location.

Myelolipomas are rare tumors that typically affect middle-aged to older adults, with a slight female predominance. They are usually discovered incidentally during imaging studies performed for other medical conditions. In most cases, myelolipomas do not require treatment unless they cause symptoms or grow large enough to pose a risk of bleeding or rupture. Surgical removal is the standard treatment for symptomatic or complicated myelolipomas.

The adrenal glands are a pair of endocrine glands that are located on top of the kidneys. Each gland has two parts: the outer cortex and the inner medulla. The adrenal cortex produces hormones such as cortisol, aldosterone, and androgens, which regulate metabolism, blood pressure, and other vital functions. The adrenal medulla produces catecholamines, including epinephrine (adrenaline) and norepinephrine (noradrenaline), which help the body respond to stress by increasing heart rate, blood pressure, and alertness.

An Adrenal Rest Tumor is a rare, benign (non-cancerous) growth that occurs in the adrenal glands. These tumors are made up of cells called "adrenal rests," which are small clusters of adrenal tissue that can be found outside of the adrenal glands.

Adrenal rest tumors are typically asymptomatic and are often discovered incidentally during imaging studies performed for other medical reasons. However, in some cases, these tumors may produce hormones such as cortisol or aldosterone, leading to symptoms associated with hormonal imbalances, such as Cushing's syndrome or Conn's syndrome.

Treatment for adrenal rest tumors typically involves surgical removal of the tumor. In cases where the tumor is producing hormones, medication may be used to manage the hormonal imbalance before and after surgery. It is important to monitor patients with adrenal rest tumors for recurrence, as these tumors can grow back over time.

Adrenalectomy is a surgical procedure in which one or both adrenal glands are removed. The adrenal glands are small, triangular-shaped glands located on top of each kidney that produce hormones such as cortisol, aldosterone, and adrenaline (epinephrine).

There are several reasons why an adrenalectomy may be necessary. For example, the procedure may be performed to treat tumors or growths on the adrenal glands, such as pheochromocytomas, which can cause high blood pressure and other symptoms. Adrenalectomy may also be recommended for patients with Cushing's syndrome, a condition in which the body is exposed to too much cortisol, or for those with adrenal cancer.

During an adrenalectomy, the surgeon makes an incision in the abdomen or back and removes the affected gland or glands. In some cases, laparoscopic surgery may be used, which involves making several small incisions and using specialized instruments to remove the gland. After the procedure, patients may need to take hormone replacement therapy to compensate for the loss of adrenal gland function.

Steroid 17-alpha-hydroxylase, also known as CYP17A1, is a cytochrome P450 enzyme that plays a crucial role in steroid hormone biosynthesis. It is located in the endoplasmic reticulum of cells in the adrenal glands and gonads. This enzyme catalyzes the 17-alpha-hydroxylation and subsequent lyase cleavage of pregnenolone and progesterone, converting them into dehydroepiandrosterone (DHEA) and androstenedione, respectively. These steroid intermediates are essential for the biosynthesis of both glucocorticoids and sex steroids, including cortisol, aldosterone, estrogens, and testosterone.

Defects in the CYP17A1 gene can lead to several disorders, such as congenital adrenal hyperplasia (CAH) due to 17-alpha-hydroxylase deficiency, which is characterized by decreased production of cortisol and sex steroids and increased mineralocorticoid levels. This condition results in sexual infantilism, electrolyte imbalances, and hypertension.

Squamous cell carcinoma is a type of skin cancer that begins in the squamous cells, which are flat, thin cells that form the outer layer of the skin (epidermis). It commonly occurs on sun-exposed areas such as the face, ears, lips, and backs of the hands. Squamous cell carcinoma can also develop in other areas of the body including the mouth, lungs, and cervix.

This type of cancer usually develops slowly and may appear as a rough or scaly patch of skin, a red, firm nodule, or a sore or ulcer that doesn't heal. While squamous cell carcinoma is not as aggressive as some other types of cancer, it can metastasize (spread) to other parts of the body if left untreated, making early detection and treatment important.

Risk factors for developing squamous cell carcinoma include prolonged exposure to ultraviolet (UV) radiation from the sun or tanning beds, fair skin, a history of sunburns, a weakened immune system, and older age. Prevention measures include protecting your skin from the sun by wearing protective clothing, using a broad-spectrum sunscreen with an SPF of at least 30, avoiding tanning beds, and getting regular skin examinations.

Adrenal cortex diseases refer to a group of conditions that affect the adrenal glands, which are small glands located on top of the kidneys. The adrenal glands consist of two parts: the outer adrenal cortex and the inner medulla. The adrenal cortex is responsible for producing hormones such as cortisol, aldosterone, and androgens that regulate various bodily functions, including metabolism, blood pressure, and sexual development.

Diseases of the adrenal cortex can result from an overproduction or underproduction of these hormones. Some common adrenal cortex diseases include:

1. Addison's disease: a condition characterized by insufficient production of hormones by the adrenal glands, leading to symptoms such as fatigue, weight loss, low blood pressure, and darkening of the skin.
2. Cushing's syndrome: a condition caused by an excess of cortisol in the body, which can result from taking high doses of corticosteroid medications or from a tumor in the pituitary gland or adrenal glands. Symptoms include weight gain, particularly around the trunk and face, thinning of the skin, easy bruising, muscle weakness, and mood changes.
3. Congenital adrenal hyperplasia: a group of inherited disorders that affect the production of hormones by the adrenal glands. Depending on the specific type of congenital adrenal hyperplasia, symptoms can range from ambiguous genitalia in newborns to precocious puberty, short stature, and infertility in older children and adults.
4. Adrenal tumors: benign or cancerous growths that develop in the adrenal glands and can cause hormonal imbalances. Symptoms depend on the type of tumor and the hormones it produces.

Treatment for adrenal cortex diseases depends on the specific condition and its underlying cause. Treatment options may include medication, surgery, or radiation therapy.

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer in adults. It originates from the hepatocytes, which are the main functional cells of the liver. This type of cancer is often associated with chronic liver diseases such as cirrhosis caused by hepatitis B or C virus infection, alcohol abuse, non-alcoholic fatty liver disease (NAFLD), and aflatoxin exposure.

The symptoms of HCC can vary but may include unexplained weight loss, lack of appetite, abdominal pain or swelling, jaundice, and fatigue. The diagnosis of HCC typically involves imaging tests such as ultrasound, CT scan, or MRI, as well as blood tests to measure alpha-fetoprotein (AFP) levels. Treatment options for Hepatocellular carcinoma depend on the stage and extent of the cancer, as well as the patient's overall health and liver function. Treatment options may include surgery, radiation therapy, chemotherapy, targeted therapy, or liver transplantation.

An adenoma is a benign (noncancerous) tumor that develops from glandular epithelial cells. These types of cells are responsible for producing and releasing fluids, such as hormones or digestive enzymes, into the surrounding tissues. Adenomas can occur in various organs and glands throughout the body, including the thyroid, pituitary, adrenal, and digestive systems.

Depending on their location, adenomas may cause different symptoms or remain asymptomatic. Some common examples of adenomas include:

1. Colorectal adenoma (also known as a polyp): These growths occur in the lining of the colon or rectum and can develop into colorectal cancer if left untreated. Regular screenings, such as colonoscopies, are essential for early detection and removal of these polyps.
2. Thyroid adenoma: This type of adenoma affects the thyroid gland and may result in an overproduction or underproduction of hormones, leading to conditions like hyperthyroidism (overactive thyroid) or hypothyroidism (underactive thyroid).
3. Pituitary adenoma: These growths occur in the pituitary gland, which is located at the base of the brain and controls various hormonal functions. Depending on their size and location, pituitary adenomas can cause vision problems, headaches, or hormonal imbalances that affect growth, reproduction, and metabolism.
4. Liver adenoma: These rare benign tumors develop in the liver and may not cause any symptoms unless they become large enough to press on surrounding organs or structures. In some cases, liver adenomas can rupture and cause internal bleeding.
5. Adrenal adenoma: These growths occur in the adrenal glands, which are located above the kidneys and produce hormones that regulate stress responses, metabolism, and blood pressure. Most adrenal adenomas are nonfunctioning, meaning they do not secrete excess hormones. However, functioning adrenal adenomas can lead to conditions like Cushing's syndrome or Conn's syndrome, depending on the type of hormone being overproduced.

It is essential to monitor and manage benign tumors like adenomas to prevent potential complications, such as rupture, bleeding, or hormonal imbalances. Treatment options may include surveillance with imaging studies, medication to manage hormonal issues, or surgical removal of the tumor in certain cases.

Feminization is a process or condition in which typically male characteristics are diminished or absent, and female characteristics become more prominent. This term is often used in the context of transgender health to describe hormone therapy that helps individuals align their physical appearance with their gender identity. The goal of feminizing hormone therapy is to promote the development of secondary sexual characteristics such as breast development, softer skin, reduced muscle mass and body hair, and fat redistribution to create a more typically female body shape. It's important to note that every individual's experience with feminization is unique, and the specific changes experienced may vary depending on factors such as age, genetics, and the duration of hormone therapy.

Adrenocortical hyperfunction, also known as Cushing's syndrome, is a condition characterized by the overproduction of cortisol hormone from the adrenal glands. The adrenal glands are located on top of the kidneys and are responsible for producing several essential hormones, including cortisol. Cortisol helps regulate metabolism, blood pressure, and the body's response to stress.

In Adrenocortical hyperfunction, the adrenal glands produce too much cortisol, leading to a range of symptoms such as weight gain, particularly around the trunk and face, thinning of the skin, easy bruising, muscle weakness, mood changes, and high blood pressure. The condition can be caused by several factors, including tumors in the pituitary gland or adrenal glands, long-term use of corticosteroid medications, or genetic disorders that affect the adrenal glands.

Treatment for Adrenocortical hyperfunction depends on the underlying cause of the condition and may include surgery to remove tumors, medication to reduce cortisol production, or radiation therapy. It is essential to diagnose and treat this condition promptly, as long-term exposure to high levels of cortisol can lead to serious health complications such as diabetes, osteoporosis, and heart disease.

17-Ketosteroids are a group of steroid compounds that contain a ketone group at the 17th carbon position in their molecular structure. They are produced as metabolic byproducts of certain hormones, such as androgens and estrogens, in the human body.

The term "17-KS" or "17-ketosteroids" is often used to refer to a class of urinary steroid metabolites that can be measured in the urine to assess adrenal and gonadal function. The measurement of 17-KS is particularly useful in monitoring patients with certain endocrine disorders, such as congenital adrenal hyperplasia or adrenal tumors.

The two major 17-KS that are routinely measured in urine are androsterone and etiocholanolone, which are derived from the metabolism of testosterone and dehydroepiandrosterone (DHEA), respectively. Other 17-KS include tetrahydrocortisone, tetrahydrocortisol, and 5-androstene-3β,17β-diol.

It's worth noting that the measurement of 17-KS has largely been replaced by more specific tests, such as the measurement of individual steroid hormones or their metabolites using mass spectrometry-based methods.

17-α-Hydroxyprogesterone is a naturally occurring hormone produced by the adrenal glands and, in smaller amounts, by the ovaries and testes. It is an intermediate in the biosynthesis of steroid hormones, including cortisol, aldosterone, and sex hormones such as testosterone and estrogen.

In a medical context, 17-α-Hydroxyprogesterone may also refer to a synthetic form of this hormone that is used in the treatment of certain medical conditions. For example, a medication called 17-alpha-hydroxyprogesterone caproate (17-OHP) is used to reduce the risk of preterm birth in women who have previously given birth prematurely. It works by suppressing uterine contractions and promoting fetal lung maturity.

It's important to note that 17-alpha-Hydroxyprogesterone should only be used under the supervision of a healthcare provider, as it can have side effects and may interact with other medications.

Li-Fraumeni Syndrome (LFS) is a rare, hereditary cancer predisposition syndrome. It is characterized by a high risk of developing multiple types of cancers throughout an individual's lifetime. The condition is caused by mutations in the TP53 gene, which plays a crucial role in suppressing tumor growth and maintaining genomic stability.

Individuals with Li-Fraumeni Syndrome have an increased risk of developing various malignancies, including:

1. Sarcomas (soft tissue and bone cancers) - most commonly occurring before the age of 45
2. Breast cancer - often diagnosed at a younger age than sporadic cases
3. Leukemias (blood cancers)
4. Brain tumors, particularly gliomas and medulloblastomas
5. Adrenocortical carcinoma (a rare type of cancer affecting the adrenal glands)
6. Other cancers such as lung, melanoma, and gastrointestinal malignancies

Li-Fraumeni Syndrome is typically inherited in an autosomal dominant manner, meaning that a person has a 50% chance of inheriting the mutated gene from an affected parent. However, de novo (new) mutations can also occur, resulting in individuals with LFS who do not have a family history of the condition.

Due to the high risk of cancer development, individuals with Li-Fraumeni Syndrome require close surveillance and early intervention strategies to manage their cancer risk effectively. Regular screenings, such as magnetic resonance imaging (MRI), computerized tomography (CT) scans, and mammograms, are often recommended for early detection and treatment of potential malignancies.

Endocrine surgical procedures refer to the surgical removal or manipulation of endocrine glands or related structures. Endocrine glands are specialized organs that produce, store, and secrete hormones directly into the bloodstream. These hormones regulate various physiological processes in the body, including metabolism, growth, development, and reproduction.

Some common endocrine surgical procedures include:

1. Thyroidectomy: Surgical removal of all or part of the thyroid gland, which is located in the neck and produces hormones regulating metabolism. Indications for thyroidectomy may include thyroid cancer, benign thyroid nodules, hyperthyroidism, and Graves' disease.
2. Parathyroidectomy: Surgical removal of one or more parathyroid glands, which are located near the thyroid gland and regulate calcium levels in the blood. Indications for parathyroidectomy may include hyperparathyroidism, parathyroid tumors, and kidney stones caused by high calcium levels.
3. Adrenalectomy: Surgical removal of one or both adrenal glands, which are located on top of the kidneys and produce hormones regulating stress response, metabolism, and blood pressure. Indications for adrenalectomy may include adrenal cancer, pheochromocytoma, Cushing's syndrome, and Conn's syndrome.
4. Pancreatectomy: Surgical removal of all or part of the pancreas, which is a gland located behind the stomach and produces hormones regulating blood sugar levels (insulin and glucagon) and enzymes for digestion. Indications for pancreatectomy may include pancreatic cancer, chronic pancreatitis, and insulinoma.
5. Neuroendocrine tumor resection: Surgical removal of neuroendocrine tumors, which are rare tumors that arise from hormone-producing cells in various organs, including the pancreas, lung, and gastrointestinal tract. Indications for neuroendocrine tumor resection may include symptoms caused by hormone excess or risk of metastasis.

These surgical procedures are complex and require specialized training and expertise. Patients should consult with a qualified surgeon to discuss the risks and benefits of each procedure and determine the best course of treatment.

Immunohistochemistry (IHC) is a technique used in pathology and laboratory medicine to identify specific proteins or antigens in tissue sections. It combines the principles of immunology and histology to detect the presence and location of these target molecules within cells and tissues. This technique utilizes antibodies that are specific to the protein or antigen of interest, which are then tagged with a detection system such as a chromogen or fluorophore. The stained tissue sections can be examined under a microscope, allowing for the visualization and analysis of the distribution and expression patterns of the target molecule in the context of the tissue architecture. Immunohistochemistry is widely used in diagnostic pathology to help identify various diseases, including cancer, infectious diseases, and immune-mediated disorders.

Aldosterone is a hormone produced by the adrenal gland. It plays a key role in regulating sodium and potassium balance and maintaining blood pressure through its effects on the kidneys. Aldosterone promotes the reabsorption of sodium ions and the excretion of potassium ions in the distal tubules and collecting ducts of the nephrons in the kidneys. This increases the osmotic pressure in the blood, which in turn leads to water retention and an increase in blood volume and blood pressure.

Aldosterone is released from the adrenal gland in response to a variety of stimuli, including angiotensin II (a peptide hormone produced as part of the renin-angiotensin-aldosterone system), potassium ions, and adrenocorticotropic hormone (ACTH) from the pituitary gland. The production of aldosterone is regulated by a negative feedback mechanism involving sodium levels in the blood. High sodium levels inhibit the release of aldosterone, while low sodium levels stimulate its release.

In addition to its role in maintaining fluid and electrolyte balance and blood pressure, aldosterone has been implicated in various pathological conditions, including hypertension, heart failure, and primary hyperaldosteronism (a condition characterized by excessive production of aldosterone).

Steroidogenic Factor 1 (SF-1 or NR5A1) is a nuclear receptor protein that functions as a transcription factor, playing a crucial role in the development and regulation of the endocrine system. It is involved in the differentiation and maintenance of steroidogenic tissues such as the adrenal glands, gonads (ovaries and testes), and the hypothalamus and pituitary glands in the brain.

SF-1 regulates the expression of genes that are essential for steroid hormone biosynthesis, including enzymes involved in the production of cortisol, aldosterone, and sex steroids (androgens, estrogens). Mutations in the SF-1 gene can lead to various disorders related to sexual development, adrenal function, and fertility.

In summary, Steroidogenic Factor 1 is a critical transcription factor that regulates the development and function of steroidogenic tissues and the biosynthesis of steroid hormones.

3-Hydroxysteroid dehydrogenases (3-HSDs) are a group of enzymes that play a crucial role in steroid hormone biosynthesis. These enzymes catalyze the conversion of 3-beta-hydroxy steroids to 3-keto steroids, which is an essential step in the production of various steroid hormones, including progesterone, cortisol, aldosterone, and sex hormones such as testosterone and estradiol.

There are several isoforms of 3-HSDs that are expressed in different tissues and have distinct substrate specificities. For instance, 3-HSD type I is primarily found in the ovary and adrenal gland, where it catalyzes the conversion of pregnenolone to progesterone and 17-hydroxyprogesterone to 17-hydroxycortisol. On the other hand, 3-HSD type II is mainly expressed in the testes, adrenal gland, and placenta, where it catalyzes the conversion of dehydroepiandrosterone (DHEA) to androstenedione and androstenedione to testosterone.

Defects in 3-HSDs can lead to various genetic disorders that affect steroid hormone production and metabolism, resulting in a range of clinical manifestations such as adrenal insufficiency, ambiguous genitalia, and sexual development disorders.

The adrenal cortex hormones are a group of steroid hormones produced and released by the outer portion (cortex) of the adrenal glands, which are located on top of each kidney. These hormones play crucial roles in regulating various physiological processes, including:

1. Glucose metabolism: Cortisol helps control blood sugar levels by increasing glucose production in the liver and reducing its uptake in peripheral tissues.
2. Protein and fat metabolism: Cortisol promotes protein breakdown and fatty acid mobilization, providing essential building blocks for energy production during stressful situations.
3. Immune response regulation: Cortisol suppresses immune function to prevent overactivation and potential damage to the body during stress.
4. Cardiovascular function: Aldosterone regulates electrolyte balance and blood pressure by promoting sodium reabsorption and potassium excretion in the kidneys.
5. Sex hormone production: The adrenal cortex produces small amounts of sex hormones, such as androgens and estrogens, which contribute to sexual development and function.
6. Growth and development: Cortisol plays a role in normal growth and development by influencing the activity of growth-promoting hormones like insulin-like growth factor 1 (IGF-1).

The main adrenal cortex hormones include:

1. Glucocorticoids: Cortisol is the primary glucocorticoid, responsible for regulating metabolism and stress response.
2. Mineralocorticoids: Aldosterone is the primary mineralocorticoid, involved in electrolyte balance and blood pressure regulation.
3. Androgens: Dehydroepiandrosterone (DHEA) and its sulfate derivative (DHEAS) are the most abundant adrenal androgens, contributing to sexual development and function.
4. Estrogens: Small amounts of estrogens are produced by the adrenal cortex, mainly in women.

Disorders related to impaired adrenal cortex hormone production or regulation can lead to various clinical manifestations, such as Addison's disease (adrenal insufficiency), Cushing's syndrome (hypercortisolism), and congenital adrenal hyperplasia (CAH).

Tumor markers are substances that can be found in the body and their presence can indicate the presence of certain types of cancer or other conditions. Biological tumor markers refer to those substances that are produced by cancer cells or by other cells in response to cancer or certain benign (non-cancerous) conditions. These markers can be found in various bodily fluids such as blood, urine, or tissue samples.

Examples of biological tumor markers include:

1. Proteins: Some tumor markers are proteins that are produced by cancer cells or by other cells in response to the presence of cancer. For example, prostate-specific antigen (PSA) is a protein produced by normal prostate cells and in higher amounts by prostate cancer cells.
2. Genetic material: Tumor markers can also include genetic material such as DNA, RNA, or microRNA that are shed by cancer cells into bodily fluids. For example, circulating tumor DNA (ctDNA) is genetic material from cancer cells that can be found in the bloodstream.
3. Metabolites: Tumor markers can also include metabolic products produced by cancer cells or by other cells in response to cancer. For example, lactate dehydrogenase (LDH) is an enzyme that is released into the bloodstream when cancer cells break down glucose for energy.

It's important to note that tumor markers are not specific to cancer and can be elevated in non-cancerous conditions as well. Therefore, they should not be used alone to diagnose cancer but rather as a tool in conjunction with other diagnostic tests and clinical evaluations.

Aromatase is a enzyme that belongs to the cytochrome P450 superfamily, and it is responsible for converting androgens into estrogens through a process called aromatization. This enzyme plays a crucial role in the steroid hormone biosynthesis pathway, particularly in females where it is primarily expressed in adipose tissue, ovaries, brain, and breast tissue.

Aromatase inhibitors are used as a treatment for estrogen receptor-positive breast cancer in postmenopausal women, as they work by blocking the activity of aromatase and reducing the levels of circulating estrogens in the body.

A cell line that is derived from tumor cells and has been adapted to grow in culture. These cell lines are often used in research to study the characteristics of cancer cells, including their growth patterns, genetic changes, and responses to various treatments. They can be established from many different types of tumors, such as carcinomas, sarcomas, and leukemias. Once established, these cell lines can be grown and maintained indefinitely in the laboratory, allowing researchers to conduct experiments and studies that would not be feasible using primary tumor cells. It is important to note that tumor cell lines may not always accurately represent the behavior of the original tumor, as they can undergo genetic changes during their time in culture.

Carcinoma in situ is a medical term used to describe the earliest stage of cancer, specifically a type of cancer that begins in the epithelial tissue, which is the tissue that lines the outer surfaces of organs and body structures. In this stage, the cancer cells are confined to the layer of cells where they first developed and have not spread beyond that layer into the surrounding tissues or organs.

Carcinoma in situ can occur in various parts of the body, including the skin, cervix, breast, lung, prostate, bladder, and other areas. It is often detected through routine screening tests, such as Pap smears for cervical cancer or mammograms for breast cancer.

While carcinoma in situ is not invasive, it can still be a serious condition because it has the potential to develop into an invasive cancer if left untreated. Treatment options for carcinoma in situ may include surgery, radiation therapy, or other forms of treatment, depending on the location and type of cancer. It is important to consult with a healthcare provider to determine the best course of action for each individual case.

Dehydroepiandrosterone (DHEA) is a steroid hormone produced by the adrenal glands. It serves as a precursor to other hormones, including androgens such as testosterone and estrogens such as estradiol. DHEA levels typically peak during early adulthood and then gradually decline with age.

DHEA has been studied for its potential effects on various health conditions, including aging, cognitive function, sexual dysfunction, and certain chronic diseases. However, the evidence supporting its use for these purposes is generally limited and inconclusive. As with any supplement or medication, it's important to consult with a healthcare provider before taking DHEA to ensure safety and effectiveness.

Steroids, also known as corticosteroids, are a type of hormone that the adrenal gland produces in your body. They have many functions, such as controlling the balance of salt and water in your body and helping to reduce inflammation. Steroids can also be synthetically produced and used as medications to treat a variety of conditions, including allergies, asthma, skin conditions, and autoimmune disorders.

Steroid medications are available in various forms, such as oral pills, injections, creams, and inhalers. They work by mimicking the effects of natural hormones produced by your body, reducing inflammation and suppressing the immune system's response to prevent or reduce symptoms. However, long-term use of steroids can have significant side effects, including weight gain, high blood pressure, osteoporosis, and increased risk of infections.

It is important to note that anabolic steroids are a different class of drugs that are sometimes abused for their muscle-building properties. These steroids are synthetic versions of the male hormone testosterone and can have serious health consequences when taken in large doses or without medical supervision.

X-ray computed tomography (CT or CAT scan) is a medical imaging method that uses computer-processed combinations of many X-ray images taken from different angles to produce cross-sectional (tomographic) images (virtual "slices") of the body. These cross-sectional images can then be used to display detailed internal views of organs, bones, and soft tissues in the body.

The term "computed tomography" is used instead of "CT scan" or "CAT scan" because the machines take a series of X-ray measurements from different angles around the body and then use a computer to process these data to create detailed images of internal structures within the body.

CT scanning is a noninvasive, painless medical test that helps physicians diagnose and treat medical conditions. CT imaging provides detailed information about many types of tissue including lung, bone, soft tissue and blood vessels. CT examinations can be performed on every part of the body for a variety of reasons including diagnosis, surgical planning, and monitoring of therapeutic responses.

In computed tomography (CT), an X-ray source and detector rotate around the patient, measuring the X-ray attenuation at many different angles. A computer uses this data to construct a cross-sectional image by the process of reconstruction. This technique is called "tomography". The term "computed" refers to the use of a computer to reconstruct the images.

CT has become an important tool in medical imaging and diagnosis, allowing radiologists and other physicians to view detailed internal images of the body. It can help identify many different medical conditions including cancer, heart disease, lung nodules, liver tumors, and internal injuries from trauma. CT is also commonly used for guiding biopsies and other minimally invasive procedures.

In summary, X-ray computed tomography (CT or CAT scan) is a medical imaging technique that uses computer-processed combinations of many X-ray images taken from different angles to produce cross-sectional images of the body. It provides detailed internal views of organs, bones, and soft tissues in the body, allowing physicians to diagnose and treat medical conditions.

Prognosis is a medical term that refers to the prediction of the likely outcome or course of a disease, including the chances of recovery or recurrence, based on the patient's symptoms, medical history, physical examination, and diagnostic tests. It is an important aspect of clinical decision-making and patient communication, as it helps doctors and patients make informed decisions about treatment options, set realistic expectations, and plan for future care.

Prognosis can be expressed in various ways, such as percentages, categories (e.g., good, fair, poor), or survival rates, depending on the nature of the disease and the available evidence. However, it is important to note that prognosis is not an exact science and may vary depending on individual factors, such as age, overall health status, and response to treatment. Therefore, it should be used as a guide rather than a definitive forecast.

Incidental findings are diagnoses or conditions that are discovered unintentionally while evaluating a patient for a different condition or symptom. These findings are not related to the primary reason for the medical examination, investigation, or procedure. They can occur in various contexts such as radiology studies, laboratory tests, or physical examinations.

Incidental findings can sometimes lead to further evaluation and management, depending on their nature and potential clinical significance. However, they also pose challenges related to communication, informed consent, and potential patient anxiety or harm. Therefore, it is essential to have clear guidelines for managing incidental findings in clinical practice.

Carcinoma, papillary is a type of cancer that begins in the cells that line the glandular structures or the lining of organs. In a papillary carcinoma, the cancerous cells grow and form small finger-like projections, called papillae, within the tumor. This type of cancer most commonly occurs in the thyroid gland, but can also be found in other organs such as the lung, breast, and kidney. Papillary carcinoma of the thyroid gland is usually slow-growing and has a good prognosis, especially when it is diagnosed at an early stage.

'Tumor cells, cultured' refers to the process of removing cancerous cells from a tumor and growing them in controlled laboratory conditions. This is typically done by isolating the tumor cells from a patient's tissue sample, then placing them in a nutrient-rich environment that promotes their growth and multiplication.

The resulting cultured tumor cells can be used for various research purposes, including the study of cancer biology, drug development, and toxicity testing. They provide a valuable tool for researchers to better understand the behavior and characteristics of cancer cells outside of the human body, which can lead to the development of more effective cancer treatments.

It is important to note that cultured tumor cells may not always behave exactly the same way as they do in the human body, so findings from cell culture studies must be validated through further research, such as animal models or clinical trials.

Antineoplastic agents, hormonal, are a class of drugs used to treat cancers that are sensitive to hormones. These agents work by interfering with the production or action of hormones in the body. They can be used to slow down or stop the growth of cancer cells and may also help to relieve symptoms caused by the spread of cancer.

Hormonal therapies can work in one of two ways: they can either block the production of hormones or prevent their action on cancer cells. For example, some hormonal therapies work by blocking the action of estrogen or testosterone, which are hormones that can stimulate the growth of certain types of cancer cells.

Examples of hormonal agents used to treat cancer include:

* Aromatase inhibitors (such as letrozole, anastrozole, and exemestane), which block the production of estrogen in postmenopausal women
* Selective estrogen receptor modulators (such as tamoxifen and raloxifene), which block the action of estrogen on cancer cells
* Luteinizing hormone-releasing hormone agonists (such as leuprolide, goserelin, and triptorelin), which block the production of testosterone in men
* Antiandrogens (such as bicalutamide, flutamide, and enzalutamide), which block the action of testosterone on cancer cells

Hormonal therapies are often used in combination with other treatments, such as surgery or radiation therapy. They may be used to shrink tumors before surgery, to kill any remaining cancer cells after surgery, or to help control the spread of cancer that cannot be removed by surgery. Hormonal therapies can also be used to relieve symptoms and improve quality of life in people with advanced cancer.

It's important to note that hormonal therapies are not effective for all types of cancer. They are most commonly used to treat breast, prostate, and endometrial cancers, which are known to be sensitive to hormones. Hormonal therapies may also be used to treat other types of cancer in certain situations.

Like all medications, hormonal therapies can have side effects. These can vary depending on the specific drug and the individual person. Common side effects of hormonal therapies include hot flashes, fatigue, mood changes, and sexual dysfunction. Some hormonal therapies can also cause more serious side effects, such as an increased risk of osteoporosis or blood clots. It's important to discuss the potential risks and benefits of hormonal therapy with a healthcare provider before starting treatment.

Neoplasm staging is a systematic process used in medicine to describe the extent of spread of a cancer, including the size and location of the original (primary) tumor and whether it has metastasized (spread) to other parts of the body. The most widely accepted system for this purpose is the TNM classification system developed by the American Joint Committee on Cancer (AJCC) and the Union for International Cancer Control (UICC).

In this system, T stands for tumor, and it describes the size and extent of the primary tumor. N stands for nodes, and it indicates whether the cancer has spread to nearby lymph nodes. M stands for metastasis, and it shows whether the cancer has spread to distant parts of the body.

Each letter is followed by a number that provides more details about the extent of the disease. For example, a T1N0M0 cancer means that the primary tumor is small and has not spread to nearby lymph nodes or distant sites. The higher the numbers, the more advanced the cancer.

Staging helps doctors determine the most appropriate treatment for each patient and estimate the patient's prognosis. It is an essential tool for communication among members of the healthcare team and for comparing outcomes of treatments in clinical trials.

Messenger RNA (mRNA) is a type of RNA (ribonucleic acid) that carries genetic information copied from DNA in the form of a series of three-base code "words," each of which specifies a particular amino acid. This information is used by the cell's machinery to construct proteins, a process known as translation. After being transcribed from DNA, mRNA travels out of the nucleus to the ribosomes in the cytoplasm where protein synthesis occurs. Once the protein has been synthesized, the mRNA may be degraded and recycled. Post-transcriptional modifications can also occur to mRNA, such as alternative splicing and addition of a 5' cap and a poly(A) tail, which can affect its stability, localization, and translation efficiency.

Retroperitoneal neoplasms refer to abnormal growths or tumors that develop in the retroperitoneal space. This is the area located behind the peritoneum, which is the membrane that lines the abdominal cavity and covers the abdominal organs. The retroperitoneal space contains several vital structures such as the kidneys, adrenal glands, pancreas, aorta, and lymphatic vessels.

Retroperitoneal neoplasms can be benign or malignant (cancerous). Malignant retroperitoneal neoplasms are often aggressive and can invade surrounding tissues and organs, leading to various complications. Common types of retroperitoneal neoplasms include lymphomas, sarcomas, and metastatic tumors from other primary sites. Symptoms may vary depending on the size and location of the tumor but can include abdominal or back pain, weight loss, and swelling in the legs. Diagnosis typically involves imaging studies such as CT scans or MRI, followed by a biopsy to determine the type and grade of the tumor. Treatment options may include surgery, radiation therapy, chemotherapy, or a combination of these approaches.

Inhibins are a group of protein hormones that play a crucial role in regulating the function of the reproductive system, specifically by inhibiting the production of follicle-stimulating hormone (FSH) in the pituitary gland. They are produced and secreted primarily by the granulosa cells in the ovaries of females and Sertoli cells in the testes of males.

Inhibins consist of two subunits, an alpha subunit, and a beta subunit, which can be further divided into two types: inhibin A and inhibin B. Inhibin A is primarily produced by the granulosa cells of developing follicles in the ovary, while inhibin B is mainly produced by the Sertoli cells in the testes.

By regulating FSH production, inhibins help control the development and maturation of ovarian follicles in females and spermatogenesis in males. Abnormal levels of inhibins have been associated with various reproductive disorders, including polycystic ovary syndrome (PCOS) and certain types of cancer.

Neoplastic gene expression regulation refers to the processes that control the production of proteins and other molecules from genes in neoplastic cells, or cells that are part of a tumor or cancer. In a normal cell, gene expression is tightly regulated to ensure that the right genes are turned on or off at the right time. However, in cancer cells, this regulation can be disrupted, leading to the overexpression or underexpression of certain genes.

Neoplastic gene expression regulation can be affected by a variety of factors, including genetic mutations, epigenetic changes, and signals from the tumor microenvironment. These changes can lead to the activation of oncogenes (genes that promote cancer growth and development) or the inactivation of tumor suppressor genes (genes that prevent cancer).

Understanding neoplastic gene expression regulation is important for developing new therapies for cancer, as targeting specific genes or pathways involved in this process can help to inhibit cancer growth and progression.

Treatment outcome is a term used to describe the result or effect of medical treatment on a patient's health status. It can be measured in various ways, such as through symptoms improvement, disease remission, reduced disability, improved quality of life, or survival rates. The treatment outcome helps healthcare providers evaluate the effectiveness of a particular treatment plan and make informed decisions about future care. It is also used in clinical research to compare the efficacy of different treatments and improve patient care.

A fatal outcome is a term used in medical context to describe a situation where a disease, injury, or illness results in the death of an individual. It is the most severe and unfortunate possible outcome of any medical condition, and is often used as a measure of the severity and prognosis of various diseases and injuries. In clinical trials and research, fatal outcome may be used as an endpoint to evaluate the effectiveness and safety of different treatments or interventions.

No FAQ available that match "adrenocortical carcinoma"

No images available that match "adrenocortical carcinoma"