Inflammatory myofibroblastic tumour

Last updated
Inflammatory myofibroblastic tumour
Other namesEpithelioid inflammatory myofibroblastic sarcoma [1]
Inflammatory myofibroblastic tumour - high mag.jpg
Micrograph of an inflammatory myofibroblastic tumour of the kidney. Kidney biopsy. H&E stain.

Inflammatory myofibroblastic tumor (IMT) is a rare neoplasm of the mesodermal cells that form the connective tissues which support virtually all of the organs and tissues of the body. [2] IMT was formerly termed inflammatory pseudotumor. [3] Currently, however, inflammatory pseudotumor designates a large and heterogeneous group of soft tissue tumors that includes inflammatory myofibroblastic tumor, plasma cell granuloma, xanthomatous pseudotumor, solitary mast cell granuloma, inflammatory fibrosarcoma, [4] pseudosarcomatous myofibroblastic proliferation, myofibroblastoma, inflammatory myofibrohistiocytic proliferation, [5] and other tumors that develop from connective tissue cells. [4] Inflammatory pseudotumour is a generic term applied to various neoplastic and non-neoplastic tissue lesions which share a common microscopic appearance consisting of spindle cells and a prominent presence of the white blood cells that populate chronic or, less commonly, acute inflamed tissues. [6] [7]

Contents

Inflammatory myofibroblastic tumor was initially regarded as a benign tumor that most often developed in the lung and less commonly in almost any organ system or tissue. Over time, however, IMT cases occurred in which the tumor spread into local tissues, metastasized to distal tissues, recurred after treatment, or consisted of neoplastic cells that had pro-malignant chromosome abnormalities. Consequently, the World Health Organization, 2013, and current literature commonly describe inflammatory myofibroblastic tumor as a neoplasm with intermediate malignant potential [7] or a rarely metastasizing neoplasm. [2] In 2020, the World Health Organization reclassified IMT as a specific tumor form in the category of intermediate (rarely metastasizing) fibroblastic and myofibroblastic tumors. [8] In all events, IMT is a rare tumor with a reported incidence in 2009 of 150–200 cases/year in the United States. [9]

IMT lesions typically consist of, and are defined by, myofibrolastic spindle cells, [7] i.e. specialized cells that are longer than wide, have a microscopic appearance that merges the appearances of fibroblasts and smooth muscle cells (see myofibroblast), occur in normal as well as tumor tissues, and in normal tissues are commonly designated fibroblasts. [10] However, the lesions in some IMF cases are dominated by sheets of epithelioid cells (which may have rounded shapes) with only a minor component of spindle cells. [11] Tumors with these characteristics are regarded as a subtype of IMT termed epithelioid inflammatory myofibroblastic sarcoma (EIMS). [3] [11] [12]

The tumors in IMT and EIMS consistently contain pro-inflammatory white blood cells and in most cases tumor cells that express highly abnormal oncogenic (cancer-causing) fusion proteins such as those that contain the active portion of anaplastic lymphoma kinase (ALK). [13] It is not clear whether this inflammation, the genetic abnormalities, or both contribute to the development of IMT but drugs blocking the activities of the fusion proteins made by these genetic abnormalities may be useful in treating the disease. [14]

Signs and symptoms

IMT was regarded as a tumor that occurs in children or young adults [13] and presented in the lung, mesentery, greater omentum or, less commonly, heart, liver, spleen, pancreas, colon, small intestine, spermatic cord, prostate, uterus, eye orbit, peripheral or central nervous system nerves, brain meninges, spinal cord, or other sites. [7] However, a more recent retrospective study of 92 patients accumulated by the Surveillance, Epidemiology, and End Results (SEER) program of the National Cancer Institute found the mean age of disease onset was 47.4 years with peak occurrences at 0 to 4, 36 to 40, and >50 years old; middle-aged individuals (41 to 64 years) represented 1/3 of all cases. In this study, the commonest sites of tumor occurrence were the lower limb and hip (22% of cases), upper limb and shoulder (12% of cases), and head, face, and neck (9% of cases). [7] Another recent study of 25 patients found the commonest sites of IMT were the abdomen (40% of cases) and lung/thoracic wall (32% of cases). [13] Individual IMT cases are also reported to present in the urinary bladder, anal canal, and parameningeal spaces (i.e. sites adjacent to the meninges such as the nasopharynx, middle ear, paranasal sinuses, infratemporal fossa and pterygopalatine fossa). [2] Apparently, the age and organ/tissue distribution of IMT various with the patient population examined: in general it can present in individuals of almost any age and in almost any organ or tissue site. IMT most commonly presents as a tumor localized to a single site but may be associated with distal metastases in up to 5% of all cases [7] or up to 10% of cases in which the tumor cells express an ALK fusion protein. [13] The tumors range in size from 1–25 cm (average 6.5 cm) with two-thirds being 1.5–6.5 cm. [7] In rare cases, the tumors have spontaneously regressed. [3] [15]

Individuals with IMT present with a wide range of symptoms (e.g. pain, swelling, a mass, organ dysfunction, etc.) depending on the tumor location(s). Up to 1/3 of these individuals have symptoms of systemic inflammation such as fever, chills, night sweats, and weight loss. [2] Rare cases of IMT have developed in individuals with: a) organizing pneumonia; b) infection by Mycobacterium avium intracellulare or Corynebacterium equi (pneumonia-causing bacteria); Campylobacter jejuni (causes gastroenteritis); Lysinibacillus sphaericus (previously termed Bacillus sphaericus, a rare cause of lung infections [16] and sepsis); [17] [18] Coxiella burneti (causes Q fever); Epstein–Barr virus (causes infectious mononucleosis and Epstein–Barr virus-associated lymphoproliferative malignant diseases); and E. coli-related occlusive phlebitis of intrahepatic veins; or c) previous abdominal surgery; trauma; ventriculoperitoneal shunt in the brain; radiation therapy; and corticosteroid usage. [19] The relationship (i.e. cause or merely association) of these disease relationships to IMT is unknown. [20]

Molecular abnormalities

The neoplastic cells in 50–60% of IMT and all cases of EIMS express an abnormal ALK protein made by a somatic recombination in the ALK gene. ALK, i.e. anaplastic lymphoma kinase (also termed protein kinase B), is produced by the ALK gene. [21] In IMT, the ALK gene has merged with a gene located at another site on the same or different chromosome to form a chimeric gene consisting of a part of the new gene and a part of the ALK gene coding for ALK's activity. [22] This chimeric gene overproduces a fusion protein with excessive ALK activity. ALK is a serine/threonine-specific protein kinase that directly or indirectly stimulates PI3K/AKT/mTOR, Ras GTPase, ERKs, Janus kinase, STAT proteins, and other cell signaling elements. Activation of these elements stimulates cell growth, proliferation, survival, and other tumor-promoting behaviors. [23] [24] As an example of this chromosomal translocation, the ALK gene located on the short or "p" arm of chromosome 2 at position 23 (notated as 2p23) merges with the CLTC gene on the long, i.e. "q" arm of chromosome 17 at position 13 (notated 17q23) to form a chimeric gene notated as t(2;17)(p23;q23). This chimeric gene makes a CLTC-ALK fusion protein with uncontrolled ALK serine/threonine-specific protein kinase activity. [14] [25] Other genes that fuse with AKT found in IMT include: TFG, DCTN1, EML4, TPM3 , [3] TPM4 , [25] ATIC [26] [27] RANBP2 [28] (most if not all RAMB2-ALK chimeric genes occur in the EMIS form of IMT [3] ), CARS1, [11] [29] and SEC31L1. [30] IMT cases may express other chimeric genes in which the active parts of ROS1 (found in 10% of IFT cases and coding for a tyrosine kinase which promotes cell growth), PDGFRB (coding for a protein that may promote the development of cancer), and NTRK (coding for a receptor tyrosine kinase that may promote the development of cancer) merge with other genes. The fusion protein products of these chimeric genes, like those of ALK fusion proteins, are overproduced, overactive, and thereby may contribute to the development of IMT. [2]

Diagnosis

Histopathologic examination of the tumors in IMT generally reveals myofibroblastic spindle cell sheets in a myxoid background (i.e. a background matrix containing gelatinous mucopolysaccharides and non-sulfated glycosaminoglycans); the matrix also contains inflammatory cells, particularly plasma cells and lymphocytes occasionally mixed with eosinophils and neutrophils. The epithelioid inflammatory myofibroblastic sarcoma subtype of IMT shows sheets of epithelioid to round cells within a myxoid (i.e. appears blue or purple compared to the normal red appearance of connective tissue when appropriately H&E stained and examined under the microscope), collagenous, or mixed myxoid-collagenous matrix, <5% spindle cells, and an inflammatory cell infiltrate that in most cases consists predominantly of neutrophils or, less often, small lymphocytes or eosinophils; plasma cells occur in only a minority of EIMS cases. The neoplastic cells in the tumors of 50% to 60% of IMT cases [2] and 100% of EIMS cases [3] express an ALK fusion protein. Other genetic abnormalities occur in these cells. Testing for the presence of the ALK fusion protein and other genetic abnormalities (see next section) can help diagnose IMT. [31]

Treatment

Many sources recommend that localized IMT be treated with total resection of all tumorous tissues. [7] [13] Localized tumor recurrences may likewise be treated by total resection. There is little support for adding radiation or systemic chemotherapy to this regimen. Tumors that are not resectable, occur in inaccessible sites, are multifocal, or have metastasized are treated with aggressive therapeutic regimens. [32]

In one retrospective study, 59 patients (all <25 years old) with IMT where treated with surgery. 31 had no residual disease post-surgery; 4 of these patients had local relapses, 3 of whom were again treated surgically and 1 with surgery plus chemotherapy. Nineteen had microscopic residual disease post-surgery. Post-surgery, 6 of these patients were treated with high-dose corticosteroids; 5 with vinblastine + methotrexate chemotherapy; 3 with inhibitors of ALK; 2 with vinorelbine + low-dose cycloheximide or Ifosfamide-based chemotherapy; and 1 with cyclophosphamide + vinchristine + actinomycin D chemotherapy. Of these 19 patients, 4 had complete responses, 8 partial responses, 5 stable disease, and 2 progressive disease. Nine patients had macroscopic disease post-surgery; 5 of these patients received vincristine + methotrexate; 2 received ALK inhibitors; and 1 each received either high-dose corticosteroids or Ifosfamide-based chemotherapy. Of these 9 patients, none had complete responses; 6 had partial responses; 1 had steady disease; and 2 had progressive disease. There were no deaths among the 59 patients. The various drug regimens showed little differences in effectiveness although patients treated with ALK inhibitors trended to have longer response times. [2] Another retrospective study evaluated the response of 17 patients (aged 22–46 years; median age 32 years) with advanced disease to Adriamycin-based chemotherapy regimens, i.e. Adriamycin alone, Adriamycin + Ifosfamide, or Adriamicin + other chemotherapy drugs. No patients had a complete response, 8 patients had partial responses, 4 patients had steady disease, and 5 patients had progressive disease. Progression-free survival and overall survival times for the group were 6.6 and 21.2 months, respectively. The study also evaluated 9 patients (aged 12–31 years, median age 16) treated with methotrexate + vinblastine, methotrexate + vinorelbine, or vinblastine + vinorelbine; 2 patients attained complete responses, 3 attained partial responses, 2 had steady disease, and 2 had progressive disease; this groups' progression-free time was not reached while its overall survival time was 83.4 months. The study concluded that the Adriamycin-based and methotrexate/vinblastine/vinorelbine regimens have a high degree of activity in IMT. Due to the low numbers of patients evaluated, no conclusions could be made on which regimen(s) were most effective. [13]

In addition to the report that compared the effect of ALK-inhibitors to other therapy regimens detailed in the previous paragraph, several reports have focused primarily on small numbers of IMT patients treated with an ALK inhibitor. The European Organisation for Research and Treatment of Cancer evaluated the effect of the ALK-inhibitor, crizotinib, on 12 ALK-positive IMT adults who had persistent and/or metastatic disease following surgical and/or drug treatment: 2 patients had complete responses, 4 had partial responses, 5 had steady disease, and none had progressive disease; 9 of these patients had at least 1 year of progression-free survival but one patient died of the disease. [33] A review of previously published IMT patients of all ages found that: 1) 4 patients (3 with unifocal, 1 with multifocal disease) without prior treatment had complete responses to crizotinin; 2) 2 patients (1 with unifocal, 1 with multifocal disease) that had persistent disease after surgery and previously treated with chemotherapy had partial responses to crizotinib while 1 patient previously treated with a corticosteroid, prednisone, continued to have progressive disease on crizotinib; and 3) 6 patients with progressive disease after surgery (due to multifocal or unifocal disease in inaccessible sites) had complete responses (2 cases), partial responses (2 cases), stable disease (1 case), or progressive disease (1 case) in response to crizotinib. Two of these crizotinib-treated patients with progressive disease had near complete responses to second generation ALK inhibitors [3] Another study reviewed 29 pediatric patients (age 15 months to 17 years) who were treated with an ALK inhibitor followed by surgical removal of the tumor (5 cases), surgical tumor removal followed by an ALK inhibitor (12 cases), or an ALK inhibitor without surgery. Twelve patients had complete responses, 14 partial responses, 2 stable disease, and 2 recurrences after finishing ALK inhibitor treatment. The latter two patients obtained complete responses to retreatment with crizotinib (1 case) or a second generation ALK inhibitor, ceritinib (1 case). [14] A study of 14 pediatric patients with metastatic or inoperable ALK-positive IMT were treated with crizotinib: 5 patients obtained complete responses, 7 partial responses, and 2 stable disease. Over the study period (2–63 months), no patient developed progressive disease. [34] Numerous Medical history studies have had similar results in treated IMT with ALK inhibitors. [35] [14] [25] [36] However, ALK inhibitors have serious side effects; in on study, crizotinib treatment was associated with pneumonia, fever of unknown cause, heart attack, sepsis, abdominal abscess, acute renal insufficiency, and the development of an abnormal EKG (i.e. QT prolongation). [33] Entrectinib, a tyrosine kinase inhibitor that is active on ROS1 and NRTK as well as AKT, has shown clinically significant activity in individual cases of patients with IMT expressing ROS1, NRTK, [37] and/or an ALK fusion proteins. [38]

See also

Related Research Articles

<span class="mw-page-title-main">Anaplastic large-cell lymphoma</span> Medical condition

Anaplastic large-cell lymphoma (ALCL) refers to a group of non-Hodgkin lymphomas in which aberrant T cells proliferate uncontrollably. Considered as a single entity, ALCL is the most common type of peripheral lymphoma and represents ~10% of all peripheral lymphomas in children. The incidence of ALCL is estimated to be 0.25 cases per 100,000 people in the United States of America. There are four distinct types of anaplastic large-cell lymphomas that on microscopic examination share certain key histopathological features and tumor marker proteins. However, the four types have very different clinical presentations, gene abnormalities, prognoses, and/or treatments.

<span class="mw-page-title-main">ETV6</span> Protein-coding gene in the species Homo sapiens

ETV6 protein is a transcription factor that in humans is encoded by the ETV6 gene. The ETV6 protein regulates the development and growth of diverse cell types, particularly those of hematological tissues. However, its gene, ETV6 frequently suffers various mutations that lead to an array of potentially lethal cancers, i.e., ETV6 is a clinically significant proto-oncogene in that it can fuse with other genes to drive the development and/or progression of certain cancers. However, ETV6 is also an anti-oncogene or tumor suppressor gene in that mutations in it that encode for a truncated and therefore inactive protein are also associated with certain types of cancers.

<span class="mw-page-title-main">Mesoblastic nephroma</span> Medical condition

Congenital mesoblastic nephroma, while rare, is the most common kidney neoplasm diagnosed in the first three months of life and accounts for 3-5% of all childhood renal neoplasms. This neoplasm is generally non-aggressive and amenable to surgical removal. However, a readily identifiable subset of these kidney tumors has a more malignant potential and is capable of causing life-threatening metastases. Congenital mesoblastic nephroma was first named as such in 1967 but was recognized decades before this as fetal renal hamartoma or leiomyomatous renal hamartoma.

<span class="mw-page-title-main">Anaplastic lymphoma kinase</span> Protein-coding gene in the species Homo sapiens

Anaplastic lymphoma kinase (ALK) also known as ALK tyrosine kinase receptor or CD246 is an enzyme that in humans is encoded by the ALK gene.

<span class="mw-page-title-main">ROS1</span> Protein-coding gene in the species Homo sapiens

Proto-oncogene tyrosine-protein kinase ROS is an enzyme that in humans is encoded by the ROS1 gene.

<span class="mw-page-title-main">Enteropathy-associated T-cell lymphoma</span> Complication of coeliac disease

Enteropathy-associated T-cell lymphoma (EATL), previously termed enteropathy-associated T-cell lymphoma, type I and at one time termed enteropathy-type T-cell lymphoma (ETTL), is a complication of coeliac disease in which a malignant T-cell lymphoma develops in areas of the small intestine affected by the disease's intense inflammation. While a relatively rare disease, it is the most common type of primary gastrointestinal T-cell lymphoma.

ETV6-NTRK3 gene fusion is the translocation of genetic material between the ETV6 gene located on the short arm of chromosome 12 at position p13.2 and the NTRK3 gene located on the long arm of chromosome 15 at position q25.3 to create the (12;15)(p13;q25) fusion gene, ETV6-NTRK3. This new gene consists of the 5' end of ETV6 fused to the 3' end of NTRK3. ETV6-NTRK3 therefore codes for a chimeric oncoprotein consisting of the helix-loop-helix (HLH) protein dimerization domain of the ETV6 protein fused to the tyrosine kinase domain of the NTRK3 protein. The ETV6 gene codes for the transcription factor protein, ETV6, which suppresses the expression of, and thereby regulates, various genes that in mice are required for normal hematopoiesis as well as the development and maintenance of the vascular network. NTRK3 codes for Tropomyosin receptor kinase C a NT-3 growth factor receptor cell surface protein that when bound to its growth factor ligand, neurotrophin-3, becomes an active tyrosine kinase that phosphorylates tyrosine residues on, and thereby stimulates, signaling proteins that promote the growth, survival, and proliferation of their parent cells. The tyrosine kinase of the ETV6-NTRK3 fusion protein is dysfunctional in that it is continuously active in phosphorylating tyrosine residues on, and thereby continuously stimulating, proteins that promote the growth, survival, and proliferation of their parent cells. In consequence, these cells take on malignant characteristics and are on the pathway of becoming cancerous. Indeed, the ETV6-NTRK3 fusion gene appears to be a critical driver of several types of cancers. It was originally identified in congenital fibrosarcoma and subsequently found in mammary secretory carcinoma, mammary analogue secretory carcinoma of salivary glands, salivary gland–type carcinoma of the thyroid, secretory carcinoma of the skin, congenital fibrosarcoma, congenital mesoblastic nephroma, rare cases of acute myelogenous leukemia, ALK-negative Inflammatory myofibroblastic tumour, cholangiocarcinoma, and radiation-induced papillary thyroid carcinoma.

<span class="mw-page-title-main">Crizotinib</span> ALK inhibitor for treatment of non-small-cell lung cancer

Crizotinib, sold under the brand name Xalkori among others, is an anti-cancer medication used for the treatment of non-small cell lung carcinoma (NSCLC). It acts as an ALK and ROS1 inhibitor.

<span class="mw-page-title-main">Carlo Gambacorti-Passerini</span> Italian oncologist and hematologist

Carlo Gambacorti-Passerini is an Italian oncologist and hematologist known for his contributions to cancer research. He is Professor of Internal Medicine and Hematology at the University of Milan Bicocca in Italy and Director of the Hematology Department at S. Gerardo Hospital, Monza, Italy. He was Senior Investigator and Head of the Oncogenic Fusion Proteins Unit at the National Cancer Institute, Milan Italy from 1990 to 2003, and Professor of Oncology and Hematology at McGill University, Montreal, Quebec, Canada, from 2004 to 2007.

<span class="mw-page-title-main">ALK inhibitor</span>

ALK inhibitors are anti-cancer drugs that act on tumours with variations of anaplastic lymphoma kinase (ALK) such as an EML4-ALK translocation. They fall under the category of tyrosine kinase inhibitors, which work by inhibiting proteins involved in the abnormal growth of tumour cells. All the current approved ALK inhibitors function by binding to the ATP pocket of the abnormal ALK protein, blocking its access to energy and deactivating it. A majority of ALK-rearranged NSCLC harbour the EML4-ALK fusion, although as of 2020, over 92 fusion partners have been discovered in ALK+ NSCLC. For each fusion partner, there can be several fusion variants depending on the position the two genes were fused at, and this may have implications on the response of the tumour and prognosis of the patient.

<span class="mw-page-title-main">ALK positive lung cancer</span> Medical condition

ALK positive lung cancer is a primary malignant lung tumor whose cells contain a characteristic abnormal configuration of DNA wherein, most frequently, the echinoderm microtubule-associated protein-like 4 (EML4) gene is fused to the anaplastic lymphoma kinase (ALK) gene. Less frequently, there will be novel translocation partners for the ALK gene, in place of EML4. This abnormal gene fusion leads to the production of a protein that appears, in many cases, to promote and maintain the malignant behavior of the cancer cells.

Targeted molecular therapy for neuroblastoma involves treatment aimed at molecular targets that have a unique expression in this form of cancer. Neuroblastoma, the second most common pediatric malignant tumor, often involves treatment through intensive chemotherapy. A number of molecular targets have been identified for the treatment of high-risk forms of this disease. Aiming treatment in this way provides a more selective way to treat the disease, decreasing the risk for toxicities that are associated with the typical treatment regimen. Treatment using these targets can supplement or replace some of the intensive chemotherapy that is used for neuroblastoma. These molecular targets of this disease include GD2, ALK, and CD133. GD2 is a target of immunotherapy, and is the most fully developed of these treatment methods, but is also associated with toxicities. ALK has more recently been discovered, and drugs in development for this target are proving to be successful in neuroblastoma treatment. The role of CD133 in neuroblastoma has also been more recently discovered and is an effective target for treatment of this disease.

<span class="mw-page-title-main">Ceritinib</span> ALK inhibitor for treatment of non-small-cell lung cancer

Ceritinib is a prescription-only drug used for the treatment of non-small cell lung cancer (NSCLC). It was developed by Novartis and received FDA approval for use in April 2014..Ceritinib is also sold under the brand name Spexib in few countries by Novartis.

ALK+ large B-cell lymphoma is a type of lymphoma. It was first reported in 1997. It is a rare, aggressive large B-cell process that shows ALK expression. It is distinct from anaplastic large cell lymphoma, a T-cell lymphoma.

<span class="mw-page-title-main">Alectinib</span> ALK inhibitor for treatment of non-small-cell lung cancer

Alectinib (INN,), sold under the brand name Alecensa, is an anticancer medication that is used to treat non-small-cell lung cancer (NSCLC). It blocks the activity of anaplastic lymphoma kinase (ALK). It is taken by mouth. It was developed by Chugai Pharmaceutical Co. Japan, which is part of the Hoffmann-La Roche group.

<span class="mw-page-title-main">Entrectinib</span> TKI inhibitor used for cancer treatment

Entrectinib, sold under the brand name Rozlytrek, is an anti-cancer medication used to treat ROS1-positive non-small cell lung cancer and NTRK fusion-positive solid tumors. It is a selective tyrosine kinase inhibitor (TKI), of the tropomyosin receptor kinases (TRK) A, B and C, C-ros oncogene 1 (ROS1) and anaplastic lymphoma kinase (ALK).

Mammary analogue secretory carcinoma (MASC), also termed MASCSG, is a salivary gland neoplasm. It is a secretory carcinoma which shares the microscopic pathologic features with other types of secretory carcinomas including mammary secretory carcinoma, secretory carcinoma of the skin, and salivary gland–type carcinoma of the thyroid. MASCSG was first described by Skálová et al. in 2010. The authors of this report found a chromosome translocation in certain salivary gland tumors, i.e. a (12;15)(p13;q25) fusion gene mutation. The other secretory carcinoma types carry this fusion gene.

Monomorphic epitheliotropic intestinal T cell lymphoma (MEITL) is an extremely rare peripheral T-cell lymphoma that involves the malignant proliferation of a type of lymphocyte, the T cell, in the gastrointestinal tract. Over time, these T cells commonly spread throughout the mucosal lining of a portion of the GI tract, lead to GI tract nodules and ulcerations, and cause symptoms such as abdominal pain, weight loss, diarrhea, obstruction, bleeding, and/or perforation.

Mature T-cell lymphoma, also called peripheral T-cell lymphoma, is a group of rare, aggressive lymphomas that develop from mature white blood cells and originate from lymphoid tissues outside of the bone marrow. Mature T-cell lymphoma is under the category of non-Hodgkin lymphoma. Mature T-cell lymphomas account for 10% to 15% of all lymphomas and is more common in Asia than in Europe and America. Its common subtypes include angioimmunoblastic T-cell lymphoma, anaplastic large cell lymphoma and peripheral T-cell lymphoma not otherwise specified. While different subtypes have variable symptoms, common symptoms include enlarged painless lymph nodes, fever, weight loss, rash and night sweats.

Mammary secretory carcinoma (MSC), also termed secretory carcinoma of the breast, is a rare form of the breast cancers. MSC usually affects women but in a significant percentage of cases also occurs in men and children. Indeed, McDvitt and Stewart first described MSC in 1966 and termed it juvenile breast carcinoma because an increased number of cases were at that time diagnosed in juvenile females. MSC is the most common form of breast cancer in children, representing 80% of childhood breast cancers, although it accounts for less than 0.15% of all breast cancers.

References

  1. "Inflammatory myofibroblastic tumor | Genetic and Rare Diseases Information Center (GARD) – an NCATS Program". rarediseases.info.nih.gov. Retrieved 28 June 2019.
  2. 1 2 3 4 5 6 7 Casanova M, Brennan B, Alaggio R, Kelsey A, Orbach D, van Noesel MM, Corradini N, Minard-Colin V, Zanetti I, Bisogno G, Gallego S, Merks JH, De Salvo GL, Ferrari A (March 2020). "Inflammatory myofibroblastic tumor: The experience of the European pediatric Soft Tissue Sarcoma Study Group (EpSSG)". European Journal of Cancer. 127: 123–129. doi:10.1016/j.ejca.2019.12.021. PMID   32007712. S2CID   211012731.
  3. 1 2 3 4 5 6 7 Theilen TM, Soerensen J, Bochennek K, Becker M, Schwabe D, Rolle U, Klingebiel T, Lehrnbecher T (April 2018). "Crizotinib in ALK+ inflammatory myofibroblastic tumors-Current experience and future perspectives". Pediatric Blood & Cancer. 65 (4): e26920. doi:10.1002/pbc.26920. PMID   29286567. S2CID   3395900.
  4. 1 2 Ajani MA, Fatunla EO, Onakpoma FA, Salami AA (2020). "Inflammatory Pseudotumor: A 20-Year Single Institutional Experience". Advanced Biomedical Research. 9: 68. doi: 10.4103/abr.abr_48_20 . PMC   8012865 . PMID   33816387.
  5. Savvidou OD, Sakellariou VI, Papakonstantinou O, Skarpidi E, Papagelopoulos PJ (2015). "Inflammatory myofibroblastic tumor of the thigh: presentation of a rare case and review of the literature". Case Reports in Orthopedics. 2015: 814241. doi: 10.1155/2015/814241 . PMC   4402203 . PMID   25945274.
  6. Gleason BC, Hornick JL (April 2008). "Inflammatory myofibroblastic tumours: where are we now?". Journal of Clinical Pathology. 61 (4): 428–37. doi: 10.1136/jcp.2007.049387 . PMID   17938159.
  7. 1 2 3 4 5 6 7 8 Fu GX, Xu CC, Yao NF, Gu JZ, Jiang HL, Han XF (July 2019). "Inflammatory myofibroblastic tumor: A demographic, clinical and therapeutic study of 92 cases". Mathematical Biosciences and Engineering. 16 (6): 6794–6804. doi: 10.3934/mbe.2019339 . PMID   31698588.
  8. Sbaraglia M, Bellan E, Dei Tos AP (April 2021). "The 2020 WHO Classification of Soft Tissue Tumours: news and perspectives". Pathologica. 113 (2): 70–84. doi:10.32074/1591-951X-213. PMC   8167394 . PMID   33179614.
  9. Webb TR, Slavish J, George RE, Look AT, Xue L, Jiang Q, Cui X, Rentrop WB, Morris SW (March 2009). "Anaplastic lymphoma kinase: role in cancer pathogenesis and small-molecule inhibitor development for therapy". Expert Review of Anticancer Therapy. 9 (3): 331–56. doi:10.1586/14737140.9.3.331. PMC   2780428 . PMID   19275511.
  10. "spindle cells - Google Search". www.google.com. Retrieved 2021-11-22.
  11. 1 2 3 Mariño-Enríquez A, Wang WL, Roy A, Lopez-Terrada D, Lazar AJ, Fletcher CD, Coffin CM, Hornick JL (January 2011). "Epithelioid inflammatory myofibroblastic sarcoma: An aggressive intra-abdominal variant of inflammatory myofibroblastic tumor with nuclear membrane or perinuclear ALK". The American Journal of Surgical Pathology. 35 (1): 135–44. doi:10.1097/PAS.0b013e318200cfd5. PMID   21164297. S2CID   40339168.
  12. Telugu RB, Prabhu AJ, Kalappurayil NB, Mathai J, Gnanamuthu BR, Manipadam MT (May 2017). "Clinicopathological Study of 18 Cases of Inflammatory Myofibroblastic Tumors with Reference to ALK-1 Expression: 5-Year Experience in a Tertiary Care Center". Journal of Pathology and Translational Medicine. 51 (3): 255–263. doi:10.4132/jptm.2017.01.12. PMC   5445201 . PMID   28415158.
  13. 1 2 3 4 5 6 Baldi GG, Brahmi M, Lo Vullo S, Cojocaru E, Mir O, Casanova M, Vincenzi B, De Pas TM, Grignani G, Pantaleo MA, Blay JY, Jones RL, Le Cesne A, Frezza AM, Gronchi A, Collini P, Dei Tos AP, Morosi C, Mariani L, Casali PG, Stacchiotti S (November 2020). "The Activity of Chemotherapy in Inflammatory Myofibroblastic Tumors: A Multicenter, European Retrospective Case Series Analysis". The Oncologist. 25 (11): e1777–e1784. doi:10.1634/theoncologist.2020-0352. PMC   7648357 . PMID   32584482.
  14. 1 2 3 4 Craig E, Wiltsie LM, Beaupin LK, Baig A, Kozielski R, Rothstein DH, Li V, Twist CJ, Barth M (February 2021). "Anaplastic lymphoma kinase inhibitor therapy in the treatment of inflammatory myofibroblastic tumors in pediatric patients: Case reports and literature review". Journal of Pediatric Surgery. 56 (12): 2364–2371. doi:10.1016/j.jpedsurg.2021.02.004. PMID   33676744. S2CID   232140059.
  15. Matsubayashi H, Uesaka K, Sasaki K, Shimada S, Takada K, Ishiwatari H, Ono H (October 2019). "A Pancreatic Inflammatory Myofibroblastic Tumor with Spontaneous Remission: A Case Report with a Literature Review". Diagnostics (Basel, Switzerland). 9 (4): 150. doi: 10.3390/diagnostics9040150 . PMC   6963339 . PMID   31627359.
  16. Isaacson P, Jacobs PH, Mackenzie AM, Mathews AW (September 1976). "Pseudotumour of the lung caused by infection with Bacillus sphaericus". Journal of Clinical Pathology. 29 (9): 806–11. doi:10.1136/jcp.29.9.806. PMC   476182 . PMID   977782.
  17. Castagnola E, Fioredda F, Barretta MA, Pescetto L, Garaventa A, Lanino E, Micalizzi C, Giacchino R, Dini G (June 2001). "Bacillus sphaericus bacteraemia in children with cancer: case reports and literature review". The Journal of Hospital Infection. 48 (2): 142–5. doi:10.1053/jhin.2001.0995. PMID   11428882.
  18. Banerjee C, Bustamante CI, Wharton R, Talley E, Wade JC (August 1988). "Bacillus infections in patients with cancer". Archives of Internal Medicine. 148 (8): 1769–74. doi:10.1001/archinte.1988.00380080059017. PMID   3401098.
  19. Karnak I, Senocak ME, Ciftci AO, Cağlar M, Bingöl-Koloğlu M, Tanyel FC, Büyükpamukçu N (June 2001). "Inflammatory myofibroblastic tumor in children: diagnosis and treatment". Journal of Pediatric Surgery. 36 (6): 908–12. doi:10.1053/jpsu.2001.23970. PMID   11381424.
  20. "Inflammatory myofibroblastic tumor | Genetic and Rare Diseases Information Center (GARD) – an NCATS Program".
  21. "AKT1 AKT serine/threonine kinase 1 [Homo sapiens (human)] - Gene - NCBI". www.ncbi.nlm.nih.gov. Retrieved 2021-11-22.
  22. Amador C, Feldman AL (March 2021). "How I Diagnose Anaplastic Large Cell Lymphoma". American Journal of Clinical Pathology. 155 (4): 479–497. doi:10.1093/ajcp/aqab012. PMID   33686426.
  23. Ducray SP, Natarajan K, Garland GD, Turner SD, Egger G (July 2019). "The Transcriptional Roles of ALK Fusion Proteins in Tumorigenesis". Cancers. 11 (8): 1074. doi: 10.3390/cancers11081074 . PMC   6721376 . PMID   31366041.
  24. Martorana F, Motta G, Pavone G, Motta L, Stella S, Vitale SR, Manzella L, Vigneri P (2021). "AKT Inhibitors: New Weapons in the Fight Against Breast Cancer?". Frontiers in Pharmacology. 12: 662232. doi: 10.3389/fphar.2021.662232 . PMC   8118639 . PMID   33995085.
  25. 1 2 3 Cao Z, Gao Q, Fu M, Ni N, Pei Y, Ou WB (February 2019). "Anaplastic lymphoma kinase fusions: Roles in cancer and therapeutic perspectives". Oncology Letters. 17 (2): 2020–2030. doi:10.3892/ol.2018.9856. PMC   6341817 . PMID   30675269.
  26. "ATIC 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase [Homo sapiens (Human)] – Gene – NCBI".
  27. Debiec-Rychter M, Marynen P, Hagemeijer A, Pauwels P (October 2003). "ALK-ATIC fusion in urinary bladder inflammatory myofibroblastic tumor". Genes, Chromosomes & Cancer. 38 (2): 187–90. doi:10.1002/gcc.10267. PMID   12939746. S2CID   40569327.
  28. Ma Z, Hill DA, Collins MH, Morris SW, Sumegi J, Zhou M, Zuppan C, Bridge JA (May 2003). "Fusion of ALK to the Ran-binding protein 2 (RANBP2) gene in inflammatory myofibroblastic tumor". Genes, Chromosomes & Cancer. 37 (1): 98–105. doi:10.1002/gcc.10177. PMID   12661011. S2CID   23427194.
  29. "SEC31A SEC31 homolog A, COPII coat complex component [Homo sapiens (Human)] – Gene – NCBI".
  30. Panagopoulos I, Nilsson T, Domanski HA, Isaksson M, Lindblom P, Mertens F, Mandahl N (March 2006). "Fusion of the SEC31L1 and ALK genes in an inflammatory myofibroblastic tumor". International Journal of Cancer. 118 (5): 1181–6. doi: 10.1002/ijc.21490 . PMID   16161041.
  31. Hou TC, Wu PS, Huang WY, Yang YT, Tan KT, Liu SH, Chen YJ, Chen SJ, Su YW (March 2020). "Over expression of CDK4 and MDM2 in a patient with recurrent ALK-negative mediastinal inflammatory myofibroblastic tumor: A case report". Medicine. 99 (12): e19577. doi:10.1097/MD.0000000000019577. PMC   7220190 . PMID   32195970.
  32. "UpToDate".
  33. 1 2 Schöffski P, Sufliarsky J, Gelderblom H, Blay JY, Strauss SJ, Stacchiotti S, Rutkowski P, Lindner LH, Leahy MG, Italiano A, Isambert N, Debiec-Rychter M, Sciot R, Van Cann T, Marréaud S, Nzokirantevye A, Collette S, Wozniak A (June 2018). "Crizotinib in patients with advanced, inoperable inflammatory myofibroblastic tumours with and without anaplastic lymphoma kinase gene alterations (European Organisation for Research and Treatment of Cancer 90101 CREATE): a multicentre, single-drug, prospective, non-randomised phase 2 trial". The Lancet. Respiratory Medicine. 6 (6): 431–441. doi:10.1016/S2213-2600(18)30116-4. PMID   29669701. S2CID   5000248.
  34. Mossé YP, Voss SD, Lim MS, Rolland D, Minard CG, Fox E, Adamson P, Wilner K, Blaney SM, Weigel BJ (October 2017). "Targeting ALK With Crizotinib in Pediatric Anaplastic Large Cell Lymphoma and Inflammatory Myofibroblastic Tumor: A Children's Oncology Group Study". Journal of Clinical Oncology. 35 (28): 3215–3221. doi:10.1200/JCO.2017.73.4830. PMC   5617123 . PMID   28787259.
  35. Mittal A, Gupta A, Dhamija E, Barwad A, Rastogi S (March 2021). "Sustained complete response on crizotinib in primary lung inflammatory myofibroblastic tumor – Case report and literature review". Monaldi Archives for Chest Disease = Archivio Monaldi per le Malattie del Torace. 91 (3). doi: 10.4081/monaldi.2021.1586 . PMID   33794589.
  36. Zhang C, Wang Z, Zhuang R, Guo X, Feng Y, Shen F, Liu W, Zhang Y, Tong H, Sun W, Liu J, Wang G, Dai C, Lu W, Zhou Y (2020). "Efficacy and Resistance of ALK Inhibitors in Two Inflammatory Myofibroblastic Tumor Patients with ALK Fusions Assessed by Whole Exome and RNA Sequencing". OncoTargets and Therapy. 13: 10335–10342. doi: 10.2147/OTT.S270481 . PMC   7568619 . PMID   33116613.
  37. Ambati SR, Slotkin EK, Chow-Maneval E, Basu EM (2018). "Entrectinib in Two Pediatric Patients With Inflammatory Myofibroblastic Tumors Harboring ROS1 or ALK Gene Fusions". JCO Precision Oncology. 2 (2): 1–6. doi:10.1200/PO.18.00095. PMC   7594679 . PMID   33134769.
  38. Bonvini P, Rossi E, Zin A, Manicone M, Vidotto R, Facchinetti A, Tombolan L, Affinita MC, Santoro L, Zamarchi R, Bisogno G (2021). "Case Report: Circulating Tumor Cells as a Response Biomarker in ALK-Positive Metastatic Inflammatory Myofibroblastic Tumor". Frontiers in Pediatrics. 9: 652583. doi: 10.3389/fped.2021.652583 . PMC   8116882 . PMID   33996693.

[1]

  1. DEMIR, Ömer Faruk, et al. Surgical treatment outcomes of pulmonary inflammatory myofibroblastic tumors. Annals of Thoracic Medicine, 2022, 17.1: 44.